AU2020405233A1 - Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment - Google Patents

Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment Download PDF

Info

Publication number
AU2020405233A1
AU2020405233A1 AU2020405233A AU2020405233A AU2020405233A1 AU 2020405233 A1 AU2020405233 A1 AU 2020405233A1 AU 2020405233 A AU2020405233 A AU 2020405233A AU 2020405233 A AU2020405233 A AU 2020405233A AU 2020405233 A1 AU2020405233 A1 AU 2020405233A1
Authority
AU
Australia
Prior art keywords
cognitive impairment
compound
disorder
pharmaceutically acceptable
cognitive
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
AU2020405233A
Inventor
John A. Butera
Emily Elizabeth Freeman
Robert Jason Herr
Jianxing Huang
Qin Jiang
Nicholas James MAYHEW
Belew Mekonnen
Hemantbhai Patel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agenebio Inc
Original Assignee
Agenebio Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Agenebio Inc filed Critical Agenebio Inc
Publication of AU2020405233A1 publication Critical patent/AU2020405233A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems

Abstract

This invention relates to benzodiazepine derivatives, compositions comprising therapeutically effective amounts of those derivatives and methods of using those derivatives or compositions in treating cognitive impairment associated with CNS disorders. It also relates to the use of an α5 -containing GABA

Description

BENZODIAZEPINE DERIVATIVES, COMPOSITIONS, AND METHODS FOR TREATING COGNITIVE IMPAIRMENT
Statement of Government Support
[0001] This invention was made with government support under Grant No. U01 AG041140, Grant No. UH2NS101856, and Grant No. UH3NS101856 awarded by the National Institutes of Health (NIH), and in particular, its National Institute on Aging (NIA) division, an agency of the United States Government. The United States Government has certain rights in the invention.
Related Applications
[0002] This application claims the benefit of and priority from United States Provisional Application 62/950,886, filed December 19, 2019, which is incorporated herein by reference in its entirety.
Field of the Invention
[0003] The invention relates to compounds, compositions and methods for treating cognitive impairment associated with central nervous system (CNS) disorders, cognitive impairment associated with brain cancers, and brain cancers in a subject in need thereof.
Background of the Invention
[0004] Cognitive ability may decline as a normal consequence of aging or as a consequence of a central nervous disorder.
[0005] For example, a significant population of elderly adults experiences a decline in cognitive ability that exceeds what is typical in normal aging. Such age-related loss of cognitive function is characterized clinically by progressive loss of memory, cognition, reasoning, and judgment. Mild Cognitive Impairment (MCI), Age- Associated Memory Impairment (AAMI), Age-Related Cognitive Decline (ARCD) or similar clinical groupings are among those related to such age-related loss of cognitive function. According to some estimates, there are more than 16 million people with AAMI in the
1 U.S. alone (Barker et al., 1995), and MCI is estimated to affect 5.5 - 7 million in the U.S. over the age of 65 (Plassman et al., 2008).
[0006] Cognitive impairment is also associated with other central nervous system (CNS) disorders, such as dementia, Alzheimer’s Disease (AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder (in particular, mania), amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
[0007] There is, therefore, a need for effective treatment of cognitive impairment associated with central nervous system (CNS) disorders and to improve cognitive function in patients diagnosed with, for example, age-related cognitive impairment, MCI, amnestic MCI, AAMI, ARCD, dementia, AD, prodromal AD, PTSD, schizophrenia or bipolar disorder (in particular, mania), amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction and similar central nervous system (CNS) disorders with cognitive impairment or at risk of developing them.
[0008] GABAA receptors (GABAA R) are pentameric assemblies from a pool of different subunits (al-6, b1-3, g1-3, d, e, p, q) that form a Cl- permeable channel that is gated by the neurotransmitter g-aminobutyric acid (GABA). Various pharmacological effects, including anxiety disorders, epilepsy, insomnia, pre-anesthetic sedation, and muscle relaxation, are mediated by different GABAA subtypes.
[0009] Various studies have demonstrated that reduced GABA signaling is linked to various CNS disorders with cognitive impairment. In particular, the a5-containing GABAA RS, which are relatively sparse in the mammalian brain, play a role in modifying learning and memory. Previous studies demonstrated a reduction of hippocampal expression of the a5 subunit of the GABAA receptor in rats with age-related cognitive decline (see International Patent Publication WO 2007/019312). Such results suggest that upregulation of a5-containing GABAA R function may be effective in the treatment of cognitive impairment associated with said CNS disorders. [0010] Thus, there is a need for positive allosteric modulators of a5-containing GABAA R that are useful in therapeutic preparations for the treatment of cognitive impairment associated with said CNS disorders.
Summary of the Invention
[0011] The present invention addresses the aforementioned need by providing a compound of formula V-a:
V-a, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and b together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
A is C, CR6, orN;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7, O, CR6 or C(R6)2;
E is N, NR7, CR6 or C(R6)2;
W is N, NR7, CR6 or C(R6)2;
X is N, NR7, O, CR6 or C(R6)2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
V is C or CR6, or when Z is C or CR6, V is C, CR6, or N; wherein R2 is independently substituted with 0-5 R’; m and n are independently integers selected from 0-4; p is an integer selected from 2-4; each occurrence of the bond “ ” is either a single bond or a double bond; each occurrence of R1, R2, R4, and R5 are each independently selected from: halogen, -R, -OR, -N02, -NCS, -CN, -CF2H, -CF3, -OCF2H -OCF3, -S1R3, - N(R)¾ -SR, -SOR, -S02R, -S02N(R)2, -SO3R, -(CR2)I-3R, -(CR2)I-3-OR, -(CR2)I-3- 0(CR2)I-3-R, -(CR2)O-3-C(0)NR(CR2)O-3R, -(CR2)O-3-C(0)NR(CR2)O- 3OR, -C(0)R, -C(0)C(0)R,
-C(0)CH2C(0)R, -C(S)R, -C(S)OR, -C(0)OR, -C(0)C(0)0R, -C(0)C(0)N(R)2, -OC(0)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)2,
-(CR2)O-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(0)OR,
-N(R)N(R)CON(R)2, -N(R)S02R, -N(R)S02N(R)2, -N(R)C(0)OR, -N(R)C(0)R, -N(R)C(S)R, -N(R)C(0)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH) N(R)¾ -C(0)N(OR)R, -C(=NOR)R, -OP(0)(OR)2, -P(0)(R)2, -P(0)(OR)2, -P(0)(H)( OR), CºC-R8, CH2CF3, and CHF3; each occurrence of R8 is -H, -(C1-C6) alkyl, -(C3-C6) cycloalkyl, -(C1-C6) alkyl-(C3-C6) cycloalkyl, -(C1-C6) alkyl-(C6-C10) aryl, -(C6-C10) aryl, -5-10 membered heteroaryl, or -(C1-C6) alkyl-5- 10 membered heteroaryl; wherein each R8 excluding -H and -(C1-C6) alkyl is independently substituted by 0-5 of -halogen, -(C1-C6) alkyl, -CF3, -OCF3, or 0-(Cl-C6) alkyl;
R3 is absent or is selected from: halogen, -R, -OR, -N02, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR,
-S02R, -S02N(R)2, -SO3R, -(CR2)I-3R, -(CR2)I-3-OR, -(CR2)O-3-C(0)NR(CR2)O-3R, -(CR2)O-3-C(0)NR(CR2)O-30R, -C(0)R, -C(0)C(0)R, -C(0)CH2C(0)R, -C(S)R, -C(S)OR, -C(0)OR, -C(0)C(0)0R, -C(0)C(0)N(R)2, -OC(0)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)¾ -(CR2)O-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(0)OR, -N(R)N(R)CON(R)2, -N(R)S02R, -N(R)S02N(R)2, -N(R)C(0)OR, -N(R)C(0)R, -N(R)C(S)R, -N(R)C(0)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH) N(R)¾ -C(0)N(OR)R, -C(=NOR)R, -OP(0)(OR)2, -P(0)(R)2, -P(0)(OR)2, -P(0)(H)( OR), CºC-R9, COOMe, COOEt, -(Cl-C6)alkyl-CºC-R10, CH2-OR10, and CH2-0- CH2-R10; wherein each of R9 is selected from -H, -(C1-C6) alkyl, -(C6-C10) aryl, -5-10 membered heteroaryl, -(C1-C6) alkyl-(C6-C10) aryl, -(C1-C6) alkyl-5-10 membered heteroaryl, - (C3-C6) cycloalkyl, -(C1-C6) alkyl-(C3-C6) cycloalkyl, -C(O)-(C6-C10) aryl, , -(C3- C6)cycloalkyl-(C6-C 10)aryl, wherein each R9 is independently substituted with 0-5 R11; wherein each occurrence of R11 is independently selected from -halogen, -CF3, -OH, - OCF3, OCHF2, -0-(Cl-C6)alkyl, -0-CH2-(C3-C6)cycloalkyl, -CN, -SCH3 -(C6-C10) aryl, -(Cl-C6)alkyl, and -5 to 10 membered heteroaryl, wherein R10is selected from -H, -(C1-C6) alkyl, -(C6-C10) aryl, -5-10 membered heteroaryl, -(C3-C6) cycloalkyl, -CH2-(C3-C6) cycloalkyl, -CH2-(C6-C10) aryl, and - CH2-5-10-membered heteroaryl, wherein each R10is independently substituted with 0-5 R’; wherein R7 is selected from -(Cl-C6)alkyl, -(C3-C6)cycloalkyl, -5 to 10 membered heteroaryl, -(C6-C10) aryl, -(C6-C10)aryl-(C1-C6)alkyl, and -5 to 10 membered heteroaryl-(Cl-C6)alkyl, and -5-10 membered heteroaryl, wherein each R7 is independently substituted with 0-5 R’; each R6 is independently -H or -(Cl-C6)alkyl; each R7 is independently -H or -(Cl-C6)alkyl; each R8 is independently -(Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’; each R is independently selected from:
H-,
(C 1 -C 12)-aliphatic-,
(C3 -C 10)-cycloalkyl-,
(C3-C10)-cycloalkenyl-,
[(C3 -C 10)-cy cloalkyl]-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkenyl]-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkyl]-0-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkenyl]-0-(C 1 -C 12)-aliphatic-,
(C6-C10)-aryl-,
(C6-C 10)-aryl-(C 1 -C 12)aliphatic-,
(C6-C 10)-aryl-O-(C 1 -C 12)aliphatic-,
(C6-C 10)-aryl-N(R’ ’ )-(C 1 -C 12)aliphatic-,
3- to 10- membered heterocyclyl-,
(3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-,
(3- to 10- membered heterocyclyl)-0-(Cl-C12)aliphatic-,
(3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
5- to 10- membered heteroaryl-,
(5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-,
(5- to 10- membered heteroaryl)-0-(Cl-C12)-aliphatic-, and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatic-; wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S; wherein each occurrence of R is independently substituted with 0-5 R’; or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3- C10)cycloalkyl, or a 3 - to 10- membered heterocyclyl; wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(0)N(R”)2, -C(0)OR”, -NO2, -NCS, -CN, -CF , -OCF3 and -N(R”)2; wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, -(Cl- C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)- alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)- alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR0, -CH2N(R°)2, -C(0)N(R°)2, -C(0)OR°, -NO2, -NCS, -CN,
-CF3, -OCF3 and -N(R°)2, wherein each occurrence of R° is independently selected from: -(Cl-C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10-membered heteroaryl-, and (C6-C10)-aryl-.
[0012] In another aspect, the present invention provides a compound of formula A: formula A, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein: Y and Z are each independently selected from C and N, wherein Y and Z cannot both be
N; each occurrence of the bond is either a single bond or a double bond; each R1 is independently halogen, -OH, or -0(C1-C6)alkyl; each R2 is -H, -OR8, -SR8, -(CH2)nOR8, -(CH2)nSR8; each of R9 is -H, (C6-C12)aryl, or 5- to 10-membered heteroaryl, wherein each R9 is substituted with 0-5 R11; each occurrence of R11 is independently selected from -halogen, -CF3, -OH, -OCF3, OCHF2, -0-(Cl-C6)alkyl, -CN, -SCH3 -(C6-C10)aryl, and -(Cl-C6)alkyl; and m and n are independently integers selected from 0-4.
[0013] In another aspect, the present invention provides a compound of formula B: formula B, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein R1, R2, R9 and m are as defined in formula A.
[0014] In another aspect, the present invention provides a compound of formula C: formula C, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein R1, R2, R9 and m are as defined in formula A.
[0015] The present invention also provides pharmaceutical compositions that comprise a compound of formulae V-a, A, B or C or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0016] In some embodiments, compounds of formula V-a are GABAA a5 receptor positive allosteric modulators. In some embodiments, compounds of formula A are GABAA a5 receptor positive allosteric modulators. In some embodiments, compounds of formula B are GABAA a5 receptor positive allosteric modulators. In some embodiments, compounds of formula C are GABAA a5 receptor positive allosteric modulators. Compounds of formula V-a, A, B and C can be used to treat the conditions described herein, such as through activity as GABAA a5 receptor positive allosteric modulators.
[0017] In another aspect of the invention, there is provided a method for treating cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the disclosure or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post- traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In another aspect of the invention, there is provided a method of preserving or improving cognitive function in a subject in need thereof, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In certain embodiments of the invention, a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof is administered every 12 or 24 hours. [0018] In another aspect of the invention, there is provided a method for treating brain cancers (including brain tumors, e.g., medulloblastomas), the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the disclosure or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In another aspect of the invention, there is provided a method of preserving or improving cognitive function in a subject suffering from brain cancers (including brain tumors, e.g., medulloblastomas), the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In certain embodiments of the invention, a compound of the disclosure or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof is administered every 12 or 24 hours.
[0019] In another aspect of the invention, there is provided a method for treating Parkinson’s disease psychosis, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In certain embodiments of the invention, a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof is administered every 12 or 24 hours.
[0020] In some embodiments, the compounds and compositions of the present invention are for use as a medicament. In some embodiments, the compounds and compositions of the present invention are for use in treating cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In some embodiments, the compounds and compositions of the present invention are for use as a medicament in treating brain cancers (including brain tumors, e.g., medulloblastomas). In some embodiments, the compounds and compositions of the present invention are for use as a medicament in treating cognitive impairment associated with brain cancers (including brain tumors, e.g., medulloblastomas). In some embodiments, the compounds and compositions of the present invention are for use as a medicament in treating Parkinson’s disease psychosis.
[0021] In some embodiments, this application provides the use of a compound or composition described herein in the preparation of a medicament for the treatment of cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In some embodiments, the compounds and compositions of the present invention are for use in the preparation of a medicament for the treatment of brain cancers (including brain tumors, e.g., medulloblastomas). In some embodiments, the compounds and compositions of the present invention are for use in the preparation of a medicament for the treatment of cognitive impairment associated with brain cancers (including brain tumors, e.g., medulloblastomas). In some embodiments, the compounds and compositions of the present invention are for use in the preparation of a medicament for the treatment of Parkinson’s disease psychosis.
Detailed Description of the Figures
[0022] Figure 1 is a graph depicting the effects of administering methyl 3,5- diphenylpyridazine-4-carboxylate on the spatial memory retention of ten aged-impaired (AI) rats in an eight-arm Radial Arm Maze (RAM) test. The black bars refer to rats treated with vehicle alone; open bars refer to rats treated with methyl 3,5-diphenylpyridazine-4- carboxylate at different doses; hatched bar refers to rats treated with the combination of TB21007 and methyl 3,5-diphenylpyridazine-4-carboxylate. [0023] Figure 2 is a graph showing the effect of methyl 3,5-diphenylpyridazine-4- carboxylate (administered intravenously) on the binding of Rol54513 in the hippocampus and cerebellum. Methyl 3,5-diphenylpyridazine-4-carboxylate blocked the binding of Rol54513 in the hippocampus but did not affect binding of Rol5413 in the cerebellum.
[0024] Figure 3 is a graph showing dose-dependent GABAA a5 receptor occupancy by methyl 3,5-diphenylpyridazine-4-carboxylate administered intravenously, with receptor occupancy determined either by the ratio between hippocampus (a region of high GABAA a5 receptor density) exposure of RO 15-4513 and cerebellum (a region with low GABAA a5 receptor density) exposure of RO 15-4513, or by using the GABAA a5 selective compound L-655,708 (10 mg/kg, i.v.) to define full occupancy.
[0025] Figure 4 is a graph showing exposure occupancy relationships for methyl 3,5- diphenylpyridazine-4-carboxylate in hippocampus. Methyl 3,5-diphenylpyridazine-4- carboxylate occupies about 32% of GABAA a5 receptors at exposures which are behaviorally active in aged-impaired rats.
[0026] Figures 5 is a graph depicting the effect of ethyl 3-methoxy-7-methyl-9H- benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on the spatial memory retention of ten aged-impaired (AI) rats in an eight-arm Radial Arm Maze (RAM) test. Figure 5 shows the effect of ethyl 3-methoxy-7-methyl-9H- benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on the spatial memory retention of ten aged-impaired (AI) rats in the RAM test, where the vehicle control was tested 3 times, and the different doses of ethyl 3-methoxy-7-methyl-9H- benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate were tested twice; In Figure 5, black bars refer to rats treated with vehicle alone and open bars refer to rats treated with ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3- d][l,4]diazepine-10-carboxylate at different doses.
[0027] Figure 6 is a graph showing the effect of ethyl 3-methoxy-7-methyl-9H- benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate (administered intravenously) on the binding of Rol54513 in the hippocampus and cerebellum. Ethyl 3- methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10- carboxylate blocked the binding of Ro 154513 in the hippocampus but did not affect binding of Rol5413 in the cerebellum. [0028] Figure 7 is a graph showing dose-dependent GABAA a5 receptor occupancy by ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine- 10-carboxylate administered intravenously, as calculated by the ratio between hippocampus (a region of high GABAAO.5 receptor density) exposure of RO 15-4513 and cerebellum (a region with low GABAAO.5 receptor density) exposure of RO 15-4513 to define full occupancy.
[0029] Figure 8(A)-(C) are graphs showing the effect of 6,6 dimethyl-3 -(3- hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one, as compared to vehicle dimethyl sulfoxide (DMSO), in aged-impaired rats using a Morris water maze behavioral task. Figure 8(A) shows the escape latency (i.e., the average time in seconds rats took to find the hidden platform in the water pool) during training in rats received 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2- benzothiophen-4(5H)-one and rats received vehicle DMSO; Figure 8(B) shows the amount of time spent in target annulus and opposite annulus by rats received 6,6 dimethyl-3 -(3- hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one and rats received vehicle DMSO; Figure 8(C) shows number of crossing in target annulus and opposite annulus by rats received 6,6 dimethyl-3 -(3 -hydroxypropyl)thio-l-(thiazol-2-yl)- 6,7-dihydro-2-benzothiophen-4(5H)-one and rats received vehicle DMSO.
Detailed Description of the Invention
Definitions
[0030] Unless otherwise defined herein, scientific and technical terms used in this application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclature used in connection with, and techniques of, chemistry, cell and tissue culture, molecular biology, cell and cancer biology, neurobiology, neurochemistry, virology, immunology, microbiology, pharmacology, genetics and protein and nucleic acid chemistry, described herein, are those well known and commonly used in the art.
[0031] The methods and techniques of the present invention are generally performed, unless otherwise indicated, according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout this specification. See, e.g. “Principles of Neural Science,” McGraw-Hill Medical, New York, N.Y. (2000); Motulsky, “Intuitive Biostatistics,” Oxford University Press, Inc. (1995); Lodish et ak, “Molecular Cell Biology, 4th ed.,” W. H. Freeman & Co., New York (2000); Griffiths et ak, “Introduction to Genetic Analysis, 7th ed.,” W. H. Freeman & Co., N.Y. (1999); and Gilbert et ak, “Developmental Biology, 6th ed.,” Sinauer Associates, Inc., Sunderland, MA (2000).
[0032] Chemistry terms used herein are used according to conventional usage in the art, as exemplified by “The McGraw-Hill Dictionary of Chemical Terms,” Parker S., Ed., McGraw-Hill, San Francisco, C.A. (1985).
[0033] All of the publications, patents and published patent applications referred to in this application are specifically incorporated by reference herein. In case of conflict, the present specification, including its specific definitions, will control.
[0034] Throughout this specification, the word “comprise” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer (or components) or group of integers (or components), but not the exclusion of any other integer (or components) or group of integers (or components).
[0035] The singular forms “a,” “an,” and “the” include the plurals unless the context clearly dictates otherwise.
[0036] The term “including” is used to mean “including but not limited to”. “Including” and “including but not limited to” are used interchangeably. [0037] The term “agent” is used herein to denote a chemical compound (such as an organic or inorganic compound (including, such as, a compound of the present invention), a mixture of chemical compounds), a biological macromolecule (such as a nucleic acid, an antibody, including parts thereof as well as humanized, chimeric and human antibodies and monoclonal antibodies, a protein or portion thereof, e.g ., a peptide, a lipid, a carbohydrate), or an extract made from biological materials such as bacteria, plants, fungi, or animal (particularly mammalian) cells or tissues. Agents include, for example, agents which are known with respect to structure, and those which are not known with respect to structure. The a5-containing GABAA receptor agonist activity of such agents may render them suitable as “therapeutic agents” in the methods and compositions of this invention. [0038] A “patient,” “subject,” or “individual” are used interchangeably and refer to either a human or a non-human animal. These terms include mammals, such as humans, primates, livestock animals (including bovine, porcine, etc.), companion animals ( e.g ., canine, feline, etc.) and rodents (e.g., mice and rats).
[0039] “Cognitive function” or “cognitive status” refers to any higher order intellectual brain process or brain state, respectively, involved in learning and/or memory including, but not limited to, attention, information acquisition, information processing, working memory, short-term memory, long-term memory, anterograde memory, retrograde memory, memory retrieval, discrimination learning, decision-making, inhibitory response control, attentional set-shifting, delayed reinforcement learning, reversal learning, the temporal integration of voluntary behavior, expressing an interest in one’s surroundings and self-care, speed of processing, reasoning and problem solving and social cognition.
[0040] In humans, cognitive function may be measured, for example and without limitation, by the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SCAG), the Buschke Selective Reminding Test (Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale- Revised (WMS-R) (Wechsler, 1997); the Benton Visual Retention Test, or the explicit 3- altemative forced choice task, or MATRICS consensus neuropsychological test battery.
See Folstein et ak, J Psychiatric Res 12: 189-98, (1975); Robbins et ah, Dementia 5: 266- 81, (1994); Rey, L'examen clinique en psychologic, (1964); Kluger et al , J Geriatr Psychiatry Neurol 12:168-79, (1999); Marquis et ak, 2002 and Masur et ak, 1994. Also see Buchanan, R.W., Keefe, R.S.E., Umbricht, D., Green, M.F., Laughren, T., and Marder, S.R. (2011), The FDA-NIMH-MATRICS guidelines for clinical trial design of cognitive- enhancing drugs: what do we know 5 years later? Schizophr. Bulk 37, 1209-1217.
[0041] In animal model systems, cognitive function may be measured in various conventional ways known in the art, including using a Morris Water Maze (MWM),
Barnes circular maze, elevated radial arm maze, T maze or any other mazes in which the animals use spatial information. Cognitive function can be assessed by reversal learning, extradimensional set shifting, conditional discrimination learning and assessments of reward expectancy. Other tests known in the art may also be used to assess cognitive function, such as novel object recognition and odor recognition tasks.
[0042] Cognitive function may also be measured using imaging techniques such as Positron Emission Tomography (PET), functional magnetic resonance imaging (fMRI), Single Photon Emission Computed Tomography (SPECT), or any other imaging technique that allows one to measure brain function. In animals, cognitive function may also be measured with electrophysiological techniques.
[0043] “Promoting” cognitive function refers to affecting impaired cognitive function so that it more closely resembles the function of a normal, unimpaired subject. Cognitive function may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activities of normal life at a level of proficiency as close as possible to a normal, unimpaired subject or an age-matched normal, unimpaired subject.
[0044] In some cases, “promoting” cognitive function in a subject affected by age-related cognitive refers to affecting impaired cognitive function so that it more closely resembles the function of an aged-matched normal, unimpaired subject, or the function of a young adult subject. Cognitive function of that subject may be promoted to any detectable degree, but in humans preferably is promoted sufficiently to allow an impaired subject to carry out daily activities of normal life at a level of proficiency close as possible to a normal, unimpaired subject or a young adult subject or an age-matched normal unimpaired subject.
[0045] “Preserving” cognitive function refers to affecting normal or impaired cognitive function such that it does not decline or does not fall below that observed in the subject upon first presentation or diagnosis, or delays such decline.
[0046] “Improving” cognitive function includes promoting cognitive function and/or preserving cognitive function in a subject.
[0047] “Cognitive impairment” refers to cognitive function in subjects that is not as robust as that expected in a normal, unimpaired subject. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in a normal, unimpaired subject. In some cases, “cognitive impairment” in subjects affected by aged-related cognitive impairment refers to cognitive function in subjects that is not as robust as that expected in an aged-matched normal, unimpaired subject, or the function of a young adult subject (i.e. subjects with mean scores for a given age in a cognitive test).
[0048] “Age-related cognitive impairment” refers to cognitive impairment in aged subjects, wherein their cognitive function is not as robust as that expected in an age- matched normal subject or as that expected in young adult subjects. In some cases, cognitive function is reduced by about 5%, about 10%, about 30%, or more, compared to cognitive function expected in an age-matched normal subject. In some cases, cognitive function is as expected in an age-matched normal subject, but reduced by about 5%, about 10%, about 30%, about 50% or more, compared to cognitive function expected in a young adult subject. Age-related impaired cognitive function may be associated with Mild Cognitive Impairment (MCI) (including amnestic MCI and non-amnestic MCI), Age- Associated Memory Impairment (AAMI), and Age-related Cognitive Decline (ARCD).
[0049] "Cognitive impairment" associated with AD or related to AD or in AD refers to cognitive function in subjects that is not as robust as that expected in subjects who have not been diagnosed AD using conventional methodologies and standards.
[0050] “Mild Cognitive Impairment” or “MCI” refers to a condition characterized by isolated memory impairment unaccompanied other cognitive abnormalities and relatively normal functional abilities. One set of criteria for a clinical characterization of MCI specifies the following characteristics: (1) memory complaint (as reported by patient, informant, or physician), (2) normal activities of daily living (ADLs), (3) normal global cognitive function, (4) abnormal memory for age (defined as scoring more than 1.5 standard deviations below the mean for a given age), and (5) absence of indicators of dementia (as defined by DSM-IV guidelines). Petersen et al., Srch. Neurol. 56: 303-308 (1999); Petersen, “Mild cognitive impairment: Aging to Alzheimer's Disease.” Oxford University Press, N.Y. (2003). The cognitive deficit in subjects with MCI may involve any cognition area or mental process including memory, language, association, attention, perception, problem solving, executive function and visuospatial skills. See , e.g. , Winbald et al., J. Intern. Med. 256:240-240, 2004; Meguro, Acta. Neurol. Taiwan. 15:55-57, 2008; Ellison et al, CNS Spectr. 13:66-72, 2008, Petersen, Semin. Neurol. 27:22-31, 2007. MCI is further subdivided into amnestic MCI (aMCI) and non-amnestic MCI, characterized by the impairment (or lack thereof) of memory in particular. MCI is defined as aMCI if memory is found to be impaired given the age and education level of the subject. If, on the other hand, the memory of the subject is found to be intact for age and education, but other non-memory cognitive domains are impaired, such as language, executive function, or visuospatial skills, MCI is defined an non-amnestic MCI. aMCI and non-amnestic MCI can both be further subdivided into single or multiple domain MCI. aMCI-single domain refers to a condition where memory, but not other cognitive areas are impaired. aMCI- multiple domain refers to a condition where memory and at least one other cognitive area are impaired. Non-amnestic MCI is single domain or multiple domain dependent on whether nor not more than one non-memory cognitive area is impaired. See , e.g. , Peterson and Negash, CNS Spectr. 13:45-53, 2008.
[0051] Diagnosis of MCI usually entails an objective assessment of cognitive impairment, which can be garnered through the use of well-established neuropsychological tests, including the Mini Mental State Examination (MMSE), the Cambridge Neuropsychological Test Automated Battery (CANTAB) and individual tests such as Rey Auditory Verbal Learning Test (AVLT), Logical Memory Subtest of the revised Wechsler Memory Scale (WMS-R) and the New York University (NYU) Paragraph Recall Test. See Folstein et ah, J Psychiatric Res 12: 189-98 (1975); Robbins et al., Dementia 5: 266-81 (1994); Kluger et ah, J Geriatric Psychiatry Neurol 12:168-79 (1999).
[0052] "Age-Associate Memory Impairment (AAMI)" refers to a decline in memory due to aging. A patient may be considered to have AAMI if he or she is at least 50 years old and meets all of the following criteria: a) The patient has noticed a decline in memory performance, b) The patient performs worse on a standard test of memory compared to young adults, c) All other obvious causes of memory decline, except normal aging, have been ruled out (in other words, the memory decline cannot be attributed to other causes such as a recent heart attack or head injury, depression, adverse reactions to medication, Alzheimer's disease, etc.).
[0053] "Age-Related Cognitive Decline (ARCD)" refers to declines in memory and cognitive abilities that are a normal consequence of aging in humans (e.g., Craik & Salthouse, 1992). This is also true in virtually all mammalian species. Age-Associated Memory Impairment refers to older persons with objective memory declines relative to their younger years, but cognitive functioning that is normal relative to their age peers (Crook et al., 1986). Age-Consistent Memory Decline is a less pejorative label which emphasizes that these are normal developmental changes (Crook, 1993; Larrabee, 1996), are not pathophysiological (Smith et al., 1991), and rarely progress to overt dementia (Youngjohn & Crook, 1993). The DSM-IV (1994) has codified the diagnostic classification of ARCD.
[0054] “Dementia” refers to a condition characterized by severe cognitive deficit that interferes in normal activities of daily living. Subjects with dementia also display other symptoms such as impaired judgment, changes in personality, disorientation, confusion, behavior changes, trouble speaking, and motor deficits. There are different types of dementias, such as Alzheimer’s disease (AD), vascular dementia, dementia with Lewy bodies, and frontotemporal dementia.
[0055] Alzheimer’s disease (AD) is characterized by memory deficits in its early phase. Later symptoms include impaired judgment, disorientation, confusion, behavior changes, trouble speaking, and motor deficits. Histologically, AD is characterized by beta-amyloid plaques and tangles of protein tau.
[0056] Vascular dementia is caused by strokes. Symptoms overlap with those of AD, but without the focus on memory impairment.
[0057] Dementia with Lewy bodies is characterized by abnormal deposits of alpha- synuclein that form inside neurons in the brain. Cognitive impairment may be similar to AD, including impairments in memory and judgment and behavior changes.
[0058] Frontotemporal dementia is characterized by gliosis, neuronal loss, superficial spongiform degeneration in the frontal cortex and/or anterior temporal lobes, and Picks’ bodies. Symptoms include changes in personality and behavior, including a decline in social skills and language expression/comprehension.
[0059] “Post-traumatic stress disorder (PTSD)” refers to an anxiety disorder characterized by an immediate or delayed response to a catastrophic event, characterized by re experiencing the trauma, psychic numbing or avoidance of stimuli associated with the trauma, and increased arousal. Re-experiencing phenomena include intrusive memories, flashbacks, nightmares, and psychological or physiological distress in response to trauma reminders. Such responses produce anxiety and can have significant impact, both chronic and acute, on a patient’s quality of life and physical and emotional health. PTSD is also associated with impaired cognitive performance, and older individuals with PTSD have greater decline in cognitive performance relative to control patients.
[0060] “Schizophrenia” refers to a chronic debilitating disorder, characterized by a spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations ( e.g ., hallucinations, delusions), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g., anhedonia, affective flattening, avolition), and cognitive impairment. While abnormalities in the brain are proposed to underlie the full spectrum of psychopathology in schizophrenia, currently available antipsychotics are largely ineffective in treating cognitive impairments in patients.
[0061] "Bipolar disorder" or "BP" or "manic depressive disorder" or "manic depressive illness" refers to a chronic psychological/mood disorder which can be characterized by significant mood changes including periods of depression and euphoric manic periods. BP may be diagnosed by a skilled physician based on personal and medical history, interview consultation and physical examinations. The term "mania" or "manic periods" or other variants refers to periods where an individual exhibit some or all of the following characteristics: racing thoughts, rapid speech, elevated levels of activity and agitation as well as an inflated sense of self-esteem, euphoria, poor judgment, insomnia, impaired concentration and aggression.
[0062] “Amyotrophic lateral sclerosis,” also known as ALS, refers to a progressive, fatal, neurodegenerative disease characterized by a degeneration of motor neurons, the nerve cells in the central nervous system that control voluntary muscle movement. ALS is also characterized by neuronal degeneration in the entorhinal cortex and hippocampus, memory deficits, and neuronal hyperexcitability in different brain areas such as the cortex.
[0063] “Cancer-therapy-related cognitive impairment” refers to cognitive impairment that develops in subjects that are treated with cancer therapies such as chemotherapy (e.g., chemobrain) and radiation. Cytotoxicity and other adverse side-effects on the brain of cancer therapies result in cognitive impairment in such functions as memory, learning and attention. [0064] Parkinson’s disease (PD) is a neurological disorder characterized by a decrease of voluntary movements. The afflicted patient has reduction of motor activity and slower voluntary movements compared to the normal individual. The patient has characteristic "mask" face, a tendency to hurry while walking, bent over posture and generalized weakness of the muscles. There is a typical "lead-pipe" rigidity of passive movements. Another important feature of the disease is the tremor of the extremities occurring at rest and decreasing during movements.
[0065] “Autism,” as used herein, refers to an autism spectrum disorder characterized by a neural development disorder leading to impaired social interaction and communication by restricted and repetitive behavior. “Autism Spectrum Disorder” refers to a group of developmental disabilities that includes: autism; Asperger syndrome; pervasive developmental disorder not otherwise specified (PDD-NOS or atypical autism); Rett syndrome; and childhood disintegrative disorder.
[0066] Mental retardation is a generalized disorder characterized by significantly impaired cognitive function and deficits in adaptive behaviors. Mental retardation is often defined as an Intelligence Quotient (IQ) score of less than 70. Inborn causes are among many underlying causes for mental retardation. The dysfunction in neuronal communication is also considered one of the underlying causes for mental retardation (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10, 207-214).
[0067] In some instances, mental retardation includes, but are not limited to, Down syndrome, velocariofacial syndrome, fetal alcohol syndrome, Fragile X syndrome, Klinefelter’s syndrome, neurofibromatosis, congenital hypothyroidism, Williams syndrome, phenylketonuria (PKU), Smith-Lemli-Opitz syndrome, Prader-Willi syndrome, Phelan-McDermid syndrome, Mowat-Wilson syndrome, ciliopathy, Lowe syndrome and siderium type X-linked mental retardation. Down syndrome is a disorder that includes a combination of birth defects, including some degree of mental retardation, characteristic facial features and, often, heart defects, increased infections, problems with vision and hearing, and other health problems. Fragile X syndrome is a prevalent form of inherited mental retardation, occurring with a frequency of 1 in 4,000 males and 1 in 8,000 females. The syndrome is also characterized by developmental delay, hyperactivity, attention deficit disorder, and autistic-like behavior. There is no effective treatment for fragile X syndrome.
[0068] Obsessive compulsive disorder ("OCD") is a mental condition that is most commonly characterized by intrusive, repetitive unwanted thoughts (obsessions) resulting in compulsive behaviors and mental acts that an individual feels driven to perform (compulsion). Current epidemiological data indicates that OCD is the fourth most common mental disorder in the United States. Some studies suggest the prevalence of OCD is between one and three percent, although the prevalence of clinically recognized OCD is much lower, suggesting that many individuals with the disorder may not be diagnosed. Patients with OCD are often diagnosed by a psychologist, psychiatrist, or psychoanalyst according to the Diagnostic and Statistical Manual of Mental Disorders, 4th edition text revision (DSM-IV-TR) (2000) diagnostic criteria that include characteristics of obsessions and compulsions.
[0069] Substance addiction ( e.g ., drug addiction, alcohol addiction) is a mental disorder. The addiction is not triggered instantaneously upon exposure to substance of abuse.
Rather, it involves multiple, complex neural adaptations that develop with different time courses ranging from hours to days to months (Kauer J. A. Nat. Rev. Neurosci. 2007, 8, 844-858). The path to addiction generally begins with the voluntary use of one or more controlled substances, such as narcotics, barbiturates, methamphetamines, alcohol, nicotine, and any of a variety of other such controlled substances. Over time, with extended use of the controlled substance(s), the voluntary ability to abstain from the controlled substance(s) is compromised due to the effects of prolonged use on brain function, and thus on behavior. As such, substance addiction generally is characterized by compulsive substance craving, seeking and use that persist even in the face of negative consequences. The cravings may represent changes in the underlying neurobiology of the patient which likely must be addressed in a meaningful way if recovery is to be obtained. Substance addiction is also characterized in many cases by withdrawal symptoms, which for some substances are life threatening (e.g., alcohol, barbiturates) and in others can result in substantial morbidity (which may include nausea, vomiting, fever, dizziness, and profuse sweating), distress, and decreased ability to obtain recovery. For example, alcoholism, also known as alcohol dependence, is one such substance addiction. Alcoholism is primarily characterized by four symptoms, which include cravings, loss of control, physical dependence and tolerance. These symptoms also may characterize addictions to other controlled substances. The craving for alcohol, as well as other controlled substances, often is as strong as the need for food or water. Thus, an alcoholic may continue to drink despite serious family, health and/or legal ramifications.
[0070] “Treating” a condition or patient refers to taking steps to obtain beneficial or desired results, including clinical results. Beneficial or desired clinical results include, but are not limited to, preventing or slowing the progression of the disease or disorder, or alleviation, amelioration, or slowing the progression, of one or more symptoms of cognitive impairment associated with CNS disorders, such as age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy - related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In some embodiments, treatment comprises preventing or slowing the progression, of a CNS disorder (such as one as described herein). In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with that CNS disorder. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive deficit. Treating age-related cognitive impairment further comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI, ARCD and AAMI) into dementia ( e.g ., AD).
[0071] “Treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject’s performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject’s cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. Treatment of cognitive impairment in humans may improve cognitive function to any detectable degree, but is preferably improved sufficiently to allow the impaired subject to carry out daily activities of normal life at the same level of proficiency as a normal, unimpaired subject. In some cases, “treating cognitive impairment” refers to taking steps to improve cognitive function in a subject with cognitive impairment so that the subject’s performance in one or more cognitive tests is improved to any detectable degree, or is prevented from further decline. Preferably, that subject’s cognitive function, after treatment of cognitive impairment, more closely resembles the function of a normal, unimpaired subject. In some cases, “treating cognitive impairment” in a subject affecting by age-related cognitive impairment refers to takings steps to improve cognitive function in the subject so that the subject’s cognitive function, after treatment of cognitive impairment, more closely resembles the function of an age-matched normal, unimpaired subject, or the function of a young adult subject.
[0072] "Administering" or "administration of' a substance, a compound or an agent to a subject can be carried out using one of a variety of methods known to those skilled in the art. For example, a compound or an agent can be administered, intravenously, arterially, intradermally, intramuscularly, intraperitoneally, intravenously, subcutaneously, ocularly, sublingually, orally (by ingestion), intranasally (by inhalation), intraspinally, intracerebrally, and transdermally (by absorption, e.g, through a skin duct). A compound or agent can also appropriately be introduced by rechargeable or biodegradable polymeric devices or other devices, e.g. , patches and pumps, or formulations, which provide for the extended, slow, or controlled release of the compound or agent. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. In some aspects, the administration includes both direct administration, including self-administration, and indirect administration, including the act of prescribing a drug.
For example, as used herein, a physician who instructs a patient to self-administer a drug, or to have the drug administered by another and/or who provides a patient with a prescription for a drug is administering the drug to the patient.
[0073] Appropriate methods of administering a substance, a compound or an agent to a subject will also depend, for example, on the age of the subject, whether the subject is active or inactive at the time of administering, whether the subject is cognitively impaired at the time of administering, the extent of the impairment, and the chemical and biological properties of the compound or agent (e.g. solubility, digestibility, bioavailability, stability and toxicity). In some embodiments, a compound or an agent is administered orally, e.g. , to a subject by ingestion, or intravenously, e.g. , to a subject by injection. In some embodiments, the orally administered compound or agent is in an extended release or slow release formulation, or administered using a device for such slow or extended release. [0074] As used herein, a “a5-containing GABAA receptor agonist,” “a5-containing GABAA R agonist” or a “GABAA a5 receptor agonist” and other variations as used herein refer to a compound that enhances the function of a5-containing GABAA receptor (GABAA R), i.e ., a compound that increase GABA-gated Cl currents. In some embodiments, a5-containing GABAA R agonist as used herein refers to a positive allosteric modulator, which potentiates the activity of GABA. a5-containing GABAA receptor agonists, suitable for use in the present invention, include the a5-containing GABAA receptor agonists of all formulas and specific a5-containing GABAA receptor agonists described herein, and their hydrates, solvates, polymorphs, salts ( e.g ., pharmaceutically acceptable salts), isomers (e.g., stereoisomers, E/Z isomers, and tautomers), and combinations thereof.
[0075] “Antipsychotic”, “antipsychotic agent”, “antipsychotic drug”, or “antipsychotic compound” refers to (1) a typical or an atypical antipsychotic; (2) an agent that is selected from dopaminergic agents, glutamatergic agents, NMDA receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) (e.g, mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), Ml muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, AMP A/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents, serotonin receptor modulators, cholinergic agents, cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, MAO B inhibitors, PDE10 inhibitors, nNOS inhibits, neurosteroids, and neurotrophic factors, alpha-7 agonists or positive allosteric modulators (PAMs)PAMs, serotonin 2C agonists; and/or (3) an agent that is useful in treating one or more signs or symptoms of schizophrenia or bipolar disorder (in particular, mania).
[0076] “Typical antipsychotics”, as used herein, refer to conventional antipsychotics, which produce antipsychotic effects as well as movement related adverse effects related to disturbances in the nigrostriatal dopamine system. These extrapy rami dal side effects (EPS) include Parkinsonism, akathisia, tardive dyskinesia and dystonia. See Baldessarini and Tarazi in Goodman & Gilman's The Pharmacological Basis of Therapeutics 10 Edition, 2001, pp. 485-520.
[0077] "Atypical antipsychotics”, as used herein, refer to antipsychotic drugs that produce antipsychotic effects with little or no EPS and include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone. "Atypical" antipsychotics differ from conventional antipsychotics in their pharmacological profiles. While conventional antipsychotics are characterized principally by D2 dopamine receptor blockade, atypical antipsychotics show antagonist effects on multiple receptors including the 5HTa and 5HTC serotonin receptors and varying degrees of receptor affinities. Atypical antipsychotic drugs are commonly referred to as serotonin/dopamine antagonists, reflecting the influential hypothesis that greater affinity for the 5HT2 receptor than for the D2 receptor underlies "atypical" antipsychotic drug action or "second generation" antipsychotic drugs. However, the atypical antipsychotics often display side effects, including, but not limited to, weight gain, diabetes ( e.g ., type II diabetes mellitus), hyperlipidemia, QTc interval prolongation, myocarditis, sexual side effects, extrapy rami dal side effects and cataract. Thus, atypical antipsychotics do not represent a homogeneous class, given their differences in the context of both alleviation of clinical symptoms and their potential for inducing side effects such as the ones listed above. Further, the common side effects of the atypical antipsychotics as described above often limit the antipsychotic doses that can be used for these agents.
[0078] Memantine is chemically known as 3,5-dimethyladamantan-l-amine or 3,5- dimethyltricyclo[3.3.1.13,7]decan-l-amine, which is an uncompetitive N-methyl-D- aspartate (NMD A) receptor antagonist with moderate affinity. The proprietary names for memantine include: Axura® and Akatinol® (Merz), Namenda® (Forest Laboratories), Ebixa® and Abixa® (Lundbeck), and Memox® (Unipharm). Memantine is approved for the treatment of moderate to severe Alzheimer's disease (AD) in the United States at a dose of up to 28 mg/day. Derivatives or analogs of memantine, which include compounds that structurally or chemically resemble memantine, are also useful in the present invention. Such derivatives or analogs of memantine include, but are not limited to those compounds disclosed in U.S. Patents Nos. 3,391,142; 4,122,193; 4,273,774; and 5,061,703; U.S. Patent Application Publication US20040087658, US20050113458, US20060205822, US20090081259, US20090124659, and US20100227852; EP Patent Application Publication EP2260839A2; EP Patent EP1682109B1; and PCT Application Publication W02005079779, all of which are incorporated herein by reference. Memantine, as used in the present invention, includes memantine and its derivatives and analogs, as well as hydrates, polymorphs, prodrugs, salts, and solvates thereof. Memantine, as used herein, also includes a composition comprising memantine or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or prodrug thereof, wherein the composition optionally further comprises at least one additional therapeutic agent (such as a therapeutic agent useful for treating a CNS disorder or cognitive impairments associated thereof). In some embodiments, the memantine composition suitable for use in the present invention comprises memantine and a second therapeutic agent that is donepezil (under the trade name Aricept).
[0079] “Acetylcholinesterase inhibitor” or “AChE-I” as used herein refers to an agent that inhibits the ability of the cholinesterase enzyme to break down the neurotransmitter acetylcholine, thereby increasing the concentration and duration of acetylcholine, mainly in brain synapses or neuromuscular junctions. AChE-Is suitable for use in this application may include, for example, the subcategories of (i) reversible non-competitive inhibitors or reversible competitive inhibitors, (ii) irreversible, and (iii) quasi-irreversible inhibitors.
[0080] The term "simultaneous administration," as used herein, means that a a5-containing GABAA receptor agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator) and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I), or their pharmaceutically acceptable salts, hydrates, solvates, or polymorphs, are administered with a time separation of no more than about 15 minutes, and in some embodiments no more than about 10 minutes. When the drugs are administered simultaneously, the a5-containing GABAA receptor agonist (e.g, an a5-containing GABAA receptor positive allosteric modulator) and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I), or their salts, hydrates, solvates, or polymorphs, may be contained in the same dosage (e.g, a unit dosage form comprising both the a5-containing GABAA receptor agonist (e.g, an a5- containing GABAA receptor positive allosteric modulator) and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I) or in discrete dosages (e.g, the a5-containing GABAA receptor agonist (e.g, an a5-containing GABAA receptor positive allosteric modulator) or its salt, hydrate, solvate, or polymorph is contained in one dosage form and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I), or its salt, hydrate, solvate, or polymorph is contained in another dosage form).
[0081] The term "sequential administration" as used herein means that the a5-containing GABAA receptor agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator) and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE- I), or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, are administered with a time separation of more than about 15 minutes, and in some embodiments more than about one hour, or up to 12-24 hours. Either the a5-containing GABAA receptor agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator) or a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I) may be administered first. The a5-containing GABAA receptor agonist (e.g, a a5- containing GABAA receptor positive allosteric modulator) and a second therapeutic agent (e.g, an antipsychotic, memantine or an AChE-I), or their salts, hydrates, solvents, or polymorphs, for sequential administration may be contained in discrete dosage forms, optionally contained in the same container or package.
[0082] A "therapeutically effective amount" of a drug or agent is an amount of a drug or an agent that, when administered to a subject will have the intended therapeutic effect, e.g. improving cognitive function in a subject, e.g, a patient having cognitive impairment associated with a CNS disorder. The full therapeutic effect does not necessarily occur by administration of one dose, and may occur only after administration of a series of doses. Thus, a therapeutically effective amount may be administered in one or more administrations. The precise effective amount needed for a subject will depend upon, for example, the subject’s size, health and age, the nature and extent of the cognitive impairment or other symptoms of the CNS disorder (such as age-related cognitive impairment, Mild Cognitive Impairment (MCI), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia , bipolar, ALS, cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction), and the therapeutics or combination of therapeutics selected for administration, and the mode of administration. The skilled worker can readily determine the effective amount for a given situation by routine experimentation.
[0083] The compounds of the present invention also include prodrugs, analogs or derivatives. The term "prodrug" is art-recognized and is intended to encompass compounds or agents which, under physiological conditions, are converted into a5- containing GABAA R positive allosteric modulators. A common method for making a prodrug is to select moieties which are hydrolyzed or metabolized under physiological conditions to provide the desired compound or agent. In other embodiments, the prodrug is converted by an enzymatic activity of the host animal to a GABAA a5 receptor positive allosteric modulator.
[0084] "Analog" is used herein to refer to a compound which functionally resembles another chemical entity, but does not share the identical chemical structure. For example, an analog is sufficiently similar to a base or parent compound such that it can substitute for the base compound in therapeutic applications, despite minor structural differences.
[0085] "Derivative" is used herein to refer to the chemical modification of a compound. Chemical modifications of a compound can include, for example, replacement of hydrogen by an alkyl, acyl, or amino group. Many other modifications are also possible.
[0086] The term "aliphatic" as used herein refers to a straight chained or branched alkyl, alkenyl or alkynyl. It is understood that alkenyl or alkynyl embodiments need at least two carbon atoms in the aliphatic chain. Aliphatic groups typically contain from 1 (or 2) to 12 carbons, such as from 1 (or 2) to 4 carbons.
[0087] The term "aryl" as used herein refers to a monocyclic or bicyclic carbocyclic aromatic ring system. Aryl as used herein includes a (C6-C12)-aryl-. For example, aryl as used herein can be a C6-C10 monocyclic or C8-C12 bicyclic carbocyclic aromatic ring system. In some embodiments, aryl as used herein can be a (C6-C10)-aryl-. Phenyl (or Ph) is an example of a monocyclic aromatic ring system. Bicyclic aromatic ring systems include systems wherein both rings are aromatic, e.g ., naphthyl, and systems wherein only one of the two rings is aromatic, e.g. , tetralin.
[0088] The term “heterocyclic” as used herein refers to a monocyclic or bicyclic non aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N,
NH, S, SO, or SO2 in a chemically stable arrangement. Heterocyclic as used herein includes a 3- to 12- membered heterocyclyl- having 1-4 heteroatoms independently selected from O, N, NH, S, SO, or SO2. For example, heterocyclic as used herein can be a 3- to 10- membered monocyclic or 8- to 12- membered bicyclic non-aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH, S, SO, or SO2 in a chemically stable arrangement. In some embodiments, heterocyclic as used herein can be a 3- to 10- membered heterocyclyl- having 1-4 heteroatoms independently selected from O, N, NH, S, SO, or SO2. In a bicyclic non-aromatic ring system embodiment of "heterocyclyl,” one or both rings may contain said heteroatom or heteroatom groups. In another bicyclic "heterocyclyl" embodiment, one of the two rings may be aromatic. In yet another heterocyclic ring system embodiment, a non-aromatic heterocyclic ring may optionally be fused to an aromatic carbocycle.
[0089] Examples of heterocyclic rings include 3-lH-benzimidazol-2-one, 3-(l-alkyl)- benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3- tetrahydrothiophenyl, 2-morpholino, 3-morpholino, 4-morpholino, 2-thiomorpholino, 3- thiomorpholino, 4-thiomorpholino, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1- tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1-piperidinyl, 2- piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5-pyrazolinyl, 1- piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thiazolidinyl, 3-thiazolidinyl, 4- thiazolidinyl, 1-imidazolidinyl, 2-imidazolidinyl, 4-imidazolidinyl, 5-imidazolidinyl, indolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, benzothiolane, benzodithiane, and l,3-dihydro-imidazol-2-one.
[0090] The term "heteroaryl" as used herein refers to a monocyclic or bicyclic aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH or S in a chemically stable arrangement. Heteroaryl as used herein includes a 5- to 12- membered heteroaryl having 1-4 heteroatoms independently selected from O, N, NH or S. In some embodiments, heteroaryl as used herein can be a 5- to 10- membered heteroaryl having 1-4 heteroatoms independently selected from O, N, NH or S. For example, heteroaryl as used herein can be a 5- to 10- membered monocyclic or 8- to 12- membered bicyclic aromatic ring system having 1 to 4 heteroatom or heteroatom groups selected from O, N, NH or S in one or both rings in a chemically stable arrangement. In such a bicyclic aromatic ring system embodiment of "heteroaryl":
- both rings are aromatic; and
- one or both rings may contain said heteroatom or heteroatom groups.
[0091] Examples of heteroaryl rings include 2-furanyl, 3-furanyl, N-imidazolyl, 2- imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5- isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2- pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g, 3 -pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g, 5-tetrazolyl), triazolyl (e.g, 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, benzofuryl, benzothiophenyl, indolyl ( e.g ., 2-indolyl), pyrazolyl ( e.g ., 2-pyrazolyl), isothiazolyl, 1,2,3- oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4- thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazinyl, 1,3,5-triazinyl, quinolinyl (e.g., 2- quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g, 1-isoquinolinyl, 3- isoquinolinyl, or 4-isoquinolinyl).
[0092] The term "cycloalkyl or cycloalkenyl" refers to a monocyclic or fused or bridged bicyclic carbocyclic ring system that is not aromatic. For example, cycloalkyl or cycloalkenyl as used herein can be a C3 -CIO monocyclic or fused or bridged C8-C12 bicyclic carbocyclic ring system that is not aromatic. Cycloalkenyl rings have one or more units of unsaturation. Preferred cycloalkyl or cycloalkenyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, norbomyl, adamantyl and decalinyl.
[0093] The term “heteroaralkyl” refers to an alkyl in which a heteroaryl group is substituted for an alkyl H atom. For example, the alkyl group is any straight chain hydrocarbon, and can include from 1 to 12 carbon atoms (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl and dodecyl), wherein said alkyl group can be substituted with any heteroaryl group, including but not limited to, 2-furanyl, 3- furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3- isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2- pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5- pyrimidinyl, pyridazinyl (e.g, 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g, 5-tetrazolyl), triazolyl (e.g, 2-triazolyl and 5-triazolyl), 2-thienyl, 3- thienyl, benzofuryl, benzothiophenyl, indolyl (e.g, 2-indolyl), pyrazolyl (e.g, 2- pyrazolyl), isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3- triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazinyl, 1,3,5-triazinyl, quinolinyl (e.g, 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g, 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl.
[0094] When a substituted moiety is described without indicating the atom via which such moiety is bonded to a substituent, then the substituent may be bonded via any appropriate atom in such moiety. For example, for a substituted 5- to 10-membered heteroaryl, a substituent on the heteroaryl can be bonded to any of the ring-forming atoms of the heteroaryl ring that are substitutable (i.e., atoms bound to one or more hydrogen atoms).
[0095] When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any of the ring-forming atoms in that ring that are substitutable (i.e., atoms bound to one or more hydrogen atoms), unless otherwise specified or otherwise implicit from the context. For example, when a R group is defined as a pyridine, and said pyridine is depicted as follows : , the pyridine ring may be bound to the benzodiazepine derivative through any one of the ring carbon atoms in the pyridine ring. As another example, when a R group is defmied as a pyrazole, and , the pyrazole ring may be bound to the benzodiazepine derivative through any one of the ring carbon atoms of the pyrazole ring, or to the sp3 N-atom.
[0096] As used herein, the carbon atom designations may have the indicated integer and any intervening integer. For example, the number of carbon atoms in a (Cl-C4)-alkyl group is 1, 2, 3, or 4. It should be understood that these designations refer to the total number of atoms in the appropriate group. For example, in a (C3-C10)-heterocyclyl the total number of carbon atoms and heteroatoms is 3 (as in aziridine), 4, 5, 6 (as in morpholine), 7, 8, 9, or 10.
[0097] "Pharmaceutically acceptable salt" is used herein to refer to an agent or a compound according to the invention that is a therapeutically active, non-toxic base and acid salt form of the compounds. The acid addition salt form of a compound that occurs in its free form as a base can be obtained by treating said free base form with an appropriate acid such as an inorganic acid, for example, a hydrohalic such as hydrochloric or hydrobromic, sulfuric, nitric, phosphoric and the like; or an organic acid, such as, for example, acetic, hydroxyacetic, propanoic, lactic, pyruvic, malonic, succinic, maleic, fumaric, malic, tartaric, citric, methanesulfonic, ethanesulfonic, benzenesulfonic, p- toluenesulfonic, cyclic, salicylic, p- aminosalicylic, pamoic and the like. See, e.g ., WO
01/062726. [0098] Compounds containing acidic protons may be converted into their therapeutically active, non-toxic base addition salt form, e. g. metal or amine salts, by treatment with appropriate organic and inorganic bases. Appropriate base salt forms include, for example, ammonium salts, alkali and earth alkaline metal salts, e. g., lithium, sodium, potassium, magnesium, calcium salts and the like, salts with organic bases, e. g. N-methyl-D- glucamine, hydrabamine salts, and salts with amino acids such as, for example, arginine, lysine and the like. Conversely, said salt forms can be converted into the free forms by treatment with an appropriate base or acid.
[0099] Compounds and their salts can be in the form of a solvate, which is included within the scope of the present invention. Such solvates include for example hydrates, alcoholates and the like. See, e.g, WO 01/062726.
[0100] As used herein, the term "hydrate" refers to a combination of water with a compound wherein the water retains its molecular state as water and is either absorbed, adsorbed or contained within a crystal lattice of the substrate compound.
[0101] As used herein, the term "polymorph" refers to different crystalline forms of the same compound and other solid state molecular forms including pseudo-polymorphs, such as hydrates (e.g, bound water present in the crystalline structure) and solvates (e.g, bound solvents other than water) of the same compound. Different crystalline polymorphs have different crystal structures due to a different packing of the molecules in the lattice. This results in a different crystal symmetry and/or unit cell parameters which directly influences its physical properties such the X-ray diffraction characteristics of crystals or powders. A different polymorph, for example, will in general diffract at a different set of angles and will give different values for the intensities. Therefore, X-ray powder diffraction can be used to identify different polymorphs, or a solid form that comprises more than one polymorph, in a reproducible and reliable way. Crystalline polymorphic forms are of interest to the pharmaceutical industry and especially to those involved in the development of suitable dosage forms. If the polymorphic form is not held constant during clinical or stability studies, the exact dosage form used or studied may not be comparable from one lot to another. It is also desirable to have processes for producing a compound with the selected polymorphic form in high purity when the compound is used in clinical studies or commercial products since Impurities present may produce undesired toxicological effects. Certain polymorphic forms may exhibit enhanced thermodynamic stability or may be more readily manufactured in high purity in large quantities, and thus are more suitable for inclusion in pharmaceutical formulations. Certain polymorphs may display other advantageous physical properties such as lack of hygroscopic tendencies, improved solubility, and enhanced rates of dissolution due to different lattice energies.
[0102] This application contemplates all the isomers of the compounds of formulae V-a, A, B and C. “Isomer” as used herein includes optical isomers (such as stereoisomers, e.g ., enantiomers and diastereoisomers), Z (zusammen) or E (entgegen) isomers, and tautomers. Many of the compounds useful in the methods and compositions of this invention have at least one stereogenic center in their structure. This stereogenic center may be present in a R or a S configuration, said R and S notation is used in correspondence with the rules described in Pure Appl. Chem. (1976), 45, 11-30. The invention also relates to all stereoisomeric forms such as enantiomeric and diastereoisomeric forms of the compounds or mixtures thereof (including all possible mixtures of stereoisomers). See, e.g. , WO 01/062726. Furthermore, certain compounds which contain alkenyl groups may exist as Z (zusammen) or E (entgegen) isomers. In each instance, the invention includes both mixture and separate individual isomers. Multiple substituents on a piperidinyl or the azepanyl ring can also stand in either cis or trans relationship to each other with respect to the plane of the piperidinyl or the azepanyl ring. Some of the compounds may also exist in tautomeric forms. Such forms, although not explicitly indicated in the formulae described herein, are intended to be included within the scope of the present invention. With respect to the methods and compositions of the present invention, reference to a compound or compounds is intended to encompass that compound in each of its possible isomeric forms and mixtures thereof unless the particular isomeric form is referred to specifically. See, e.g., WO 01/062726.
[0103] The compounds of the invention enhance the function of a5-containing GABAA R, i.e., they are a5-containing GABAA R agonists (e.g, a5-containing GABAA receptor positive allosteric modulators) and are capable of increasing GABA-gated CT currents.
[0104] The invention further provides pharmaceutical compositions comprising one or more compounds of the invention together with a pharmaceutically acceptable carrier or excipient. In some embodiments, the pharmaceutical compositions of this application may further comprise a second therapeutic agent, such as an antipsychotic, memantine or an AChE-I. [0105] The invention further provides methods for treating cognitive impairment associated with said CNS disorders that are responsive to positive allosteric modulators of a5-containing GABAA receptor, e.g, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome,, compulsive behavior, and substance addiction. In certain embodiments, the method is a method of treating the age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction. In certain embodiments, treatment comprises preventing or slowing the progression of a CNS disorder as described herein (such as those described herein). In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with the CNS disorder. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive deficit. In another aspect of the invention, there is provided a method of preserving or improving cognitive function in a subject in need thereof, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0106] The various CNS disorders with cognitive impairment (e.g, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy - related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome,, compulsive behavior, and substance addiction) may have a variety of etiologies. However, the symptom of cognitive impairment in each of the above-mentioned disorders may have overlapping causes. Thus, a composition or method of treatment that treats cognitive impairment in one CNS disorder may also treat cognitive impairment in another.
Benzodiazepine Derivatives
[0107] The present disclosure provides compound of formula V-a:
V-a, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein:
U and the two carbon atoms designated by a and b together form a 5- or 6- membered aromatic ring having 0-2 nitrogen atoms;
A is C, CR6, orN;
B and F are each independently selected from C, CR6, and N, wherein B and F cannot both be N;
D is N, NR7, O, CR6 or C(R6)2;
E is N, NR7, CR6 or C(R6)2;
W is N, NR7, CR6 or C(R6)2;
X is N, NR7, O, CR6 or C(R6)2;
Y and Z are each independently selected from C, CR6, and N, wherein Y and Z cannot both be N;
V is C or CR6, or when Z is C or CR6, V is C, CR6, or N; in R2 is independently substituted with 0-5 R’; m and n are independently integers selected from 0-4; p is an integer selected from 2-4; each occurrence of the bond “ ” is either a single bond or a double bond; each occurrence of R1, R2, R4, and R5 are each independently selected from: halogen, -R, -OR, -N02, -NCS, -CN, -CF2H, -CF3, -OCF2H -OCF3, -S1R3, - N(R)¾ -SR, -SOR, -S02R, -S02N(R)2, -SO3R, -(CR2)I-3R, -(CR2)I-3-OR, -(CR2)I-3- 0(CR2)I-3-R, -(CR2)O-3-C(0)NR(CR2)O-3R, -(CR2)O-3-C(0)NR(CR2)O- 3OR, -C(0)R, -C(0)C(0)R,
-C(0)CH2C(0)R, -C(S)R, -C(S)OR, -C(0)OR, -C(0)C(0)0R, -C(0)C(0)N(R)2, -OC(0)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)2,
-(CR2)O-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(0)OR,
-N(R)N(R)CON(R)2, -N(R)S02R, -N(R)S02N(R)2, -N(R)C(0)OR, -N(R)C(0)R, -N(R)C(S)R, -N(R)C(0)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH) N(R)¾ -C(0)N(OR)R, -C(=NOR)R, -OP(0)(OR)2, -P(0)(R)2, -P(0)(OR)2, -P(0)(H)( OR), CºC-R8, CH2CF3, and CHF3, and in particular, in some aspects of the invention, at least one of R1, R2, R4 and R5 is -OCF2H; each occurrence of R8 is -H, -(C1-C6) alkyl, -(C3-C6) cycloalkyl, -(C1-C6) alkyl-(C3-C6) cycloalkyl, -(C1-C6) alkyl-(C6-C10) aryl, -(C6-C10) aryl, -5-10 membered heteroaryl, or -(C1-C6) alkyl-5- 10 membered heteroaryl; wherein each R8 excluding -H and -(C1-C6) alkyl is independently substituted by 0-5 of -halogen, -(C1-C6) alkyl, -CF3, -OCF3, or 0-(Cl-C6) alkyl;
R3 is absent or is selected from: halogen, -R, -OR, -N02, -NCS, -CN, -CF3, -OCF3, -SiR3, -N(R)2, -SR, -SOR,
-S02R, -S02N(R)2, -SO3R, -(CR2)I-3R, -(CR2)I-3-OR, -(CR2)O-3-C(0)NR(CR2)O-3R, -(CR2)O-3-C(0)NR(CR2)O-30R, -C(0)R, -C(0)C(0)R, -C(0)CH2C(0)R, -C(S)R, -C(S)OR, -C(0)OR, -C(0)C(0)0R, -C(0)C(0)N(R)2, -OC(0)R, -C(0)N(R)2, -OC(0)N(R)2, -C(S)N(R)¾ -(CR2)O-3NHC(0)R, -N(R)N(R)COR, -N(R)N(R)C(0)OR, -N(R)N(R)CON(R)2, -N(R)S02R, -N(R)S02N(R)2, -N(R)C(0)OR, -N(R)C(0)R, -N(R)C(S)R, -N(R)C(0)N(R)2, -N(R)C(S)N(R)2, -N(COR)COR, -N(OR)R, -C(=NH) N(R)¾ -C(0)N(OR)R, -C(=NOR)R, -OP(0)(OR)2, -P(0)(R)2, -P(0)(OR)2, -P(0)(H)( OR), CºC-R9, COOMe, COOEt, -(Cl-C6)alkyl-CºC-R10, CH2-OR10, and CH2-0- CH2-R10; wherein each of R9 is selected from -H, -(C1-C6) alkyl, -(C6-C10) aryl, -5-10 membered heteroaryl, -(C1-C6) alkyl-(C6-C10) aryl, -(C1-C6) alkyl-5-10 membered heteroaryl, - (C3-C6) cycloalkyl, -(C1-C6) alkyl-(C3-C6) cycloalkyl, -C(O)-(C6-C10) aryl, -(C3- C6)cycloalkyl-(C6-C 10)aryl, and in particular, in some aspects of the invention. R9 is selected from -(C3-C6)cycloalkyl-
(C6-C10)aryl, wherein each R is independently substituted with 0-5 R ; wherein each occurrence of R11 is independently selected from -halogen, -CF3, -OH, - OCF3, OCHF2, -0-(Cl-C6)alkyl, -0-CH2-(C3-C6)cycloalkyl, -CN, -SCH3 -(C6-C10) aryl, -(Cl-C6)alkyl, and -5 to 10 membered heteroaryl, and in particular, in some aspects of the invention, R11 is independently selected from -halogen, -OH, -OCHF2, - 0-(Cl-C6)alkyl, -0-CH2-(C3-C6)cycloalkyl, -CN, and -SCH ; wherein R10is selected from -H, -(C1-C6) alkyl, -(C6-C10) aryl, -5-10 membered heteroaryl, -(C3-C6) cycloalkyl, -CH2-(C3-C6) cycloalkyl, -CH2-(C6-C10) aryl, and - CH2-5-10-membered heteroaryl, wherein each R10is independently substituted with 0-5 R’; wherein R7 is selected from -(Cl-C6)alkyl, -(C3-C6)cycloalkyl, -5 to 10 membered heteroaryl, -(C6-C10) aryl, -(C6-C10)aryl-(C1-C6)alkyl, and -5 to 10 membered heteroaryl-(Cl-C6)alkyl, and -5-10 membered heteroaryl, wherein each R7 is independently substituted with 0-5 R’; each R6 is independently -H or -(Cl-C6)alkyl; each R7 is independently -H or -(Cl-C6)alkyl; each R8 is independently -(Cl-C6)alkyl, -(C3-C10)-cycloalkyl, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R8 is independently substituted with 0-5 R’; each R10 is independently -(C3-C10)-cycloalkyl, 3- to 10- membered heterocyclyl-, (C6-C10)-aryl, or 5- to 10- membered heteroaryl, wherein each occurrence of R10 is independently substituted with 0-5 R’; each R is independently selected from:
H-,
(C 1 -C 12)-aliphatic-,
(C3 -C 10)-cycloalkyl-,
(C3-C10)-cycloalkenyl-,
[(C3 -C 10)-cy cloalkyl]-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkenyl]-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkyl]-0-(C 1 -C 12)-aliphatic-,
[(C3 -C 10)-cy cloalkenyl]-0-(C 1 -C 12)-aliphatic-,
(C6-C10)-aryl-,
(C6-C 10)-aryl-(C 1 -C 12)aliphatic-,
(C6-C 10)-aryl-O-(C 1 -C 12)aliphatic-,
(C6-C 10)-aryl-N(R’ ’ )-(C 1 -C 12)aliphatic-,
3- to 10- membered heterocyclyl-,
(3- to 10- membered heterocyclyl)-(Cl-C12)aliphatic-,
(3- to 10- membered heterocyclyl)-0-(Cl-C12)aliphatic-,
(3- to 10- membered heterocyclyl)-N(R”)-(Cl-C12)aliphatic-,
5- to 10- membered heteroaryl-,
(5- to 10- membered heteroaryl)-(Cl-C12)-aliphatic-,
(5- to 10- membered heteroaryl)-0-(Cl-C12)-aliphatic-, and (5- to 10- membered heteroaryl)-N(R”)-(Cl-C12)-aliphatic-; wherein said heterocyclyl has 1-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, and said heteroaryl has 1-4 heteroatoms independently selected from N, NH, O, and S; wherein each occurrence of R is independently substituted with 0-5 R’; or when two R groups bound to the same atom, the two R groups may be taken together with the atom to which they are bound to form a 3- to 10-membered aromatic or non aromatic ring having 0-4 heteroatoms independently selected from N, NH, O, S, SO, and SO2, wherein said ring is optionally substituted with 0-5 R’, and wherein said ring is optionally fused to a (C6-C10)aryl, 5- to 10- membered heteroaryl, (C3- C10)cycloalkyl, or a 3 - to 10- membered heterocyclyl; wherein each occurrence of R’ is independently selected from halogen, -R”, -OR”, oxo, -CH2OR”, -CH2NR”2, -C(0)N(R”)2, -C(0)OR”, -N02, -NCS, -CN, -CF3, -0CF3 and -N(R”)2; wherein each occurrence of R” is independently selected from H, -(Cl-C6)-alkyl, -(Cl- C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10- membered heteroaryl-, (C6-C10)-aryl-, (5- to 10- membered heteroaryl)-(Cl-C6)- alkyl-, (C6-C10)-aryl-(Cl-C6)-alkyl-, (5- to 10- membered heteroaryl)-0-(Cl-C6)- alkyl-, and (C6-C10)-aryl-O-(Cl-C6)-alkyl-, wherein each occurrence of R” is independently substituted with 0-3 substituents selected from: halogen, -R°, -OR0, oxo, -CH2OR°, -CH2N(R°)2, -C(0)N(R°)2, -C(0)OR°, -N02, -NCS, -CN,
-CF3, -OCF3 and -N(R°)2, wherein each occurrence of R° is independently selected from: -(Cl-C6)-aliphatic, (C3-C6)-cycloalkyl, 3- to 6- membered heterocyclyl, 5- to 10-membered heteroaryl-, and (C6-C10)-aryl-.
[0108] In some embodiments, the compound of formula V-a has a structure according to formula A:
A, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein:
Y and Z are each independently selected from C and N, wherein Y and Z cannot both be
N; each occurrence of the bond “ ” is either a single bond or a double bond; each R1 is independently halogen, -OH, or -0(C1-C6)alkyl; each R2 is -H, -OR8, -SR8, -(CH2)nOR8, -(CH2)nSR8; each of R9 is -H, (C6-C12)aryl, or 5- to 10-membered heteroaryl, wherein each R9 is substituted with 0-5 R11; each occurrence of R11 is independently selected from -halogen, -CF3, -OH, -OCF3, OCHF2, -0-(Cl-C6)alkyl, -CN, -SCH3 -(C6-C10)aryl, and -(Cl-C6)alkyl; and m and n are independently integers selected from 0-4.
[0109] In some embodiments, the disclosure is directed to a compound having a structure according to formula A, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein: each R1 is halogen or -OMe; each R2 is -H or -CH2OMe; each wherein each R9 is substituted with 0-5 R11; and each occurrence of R11 is independently selected from -halogen, -CF3, -OH, -OCF3, OCHF2, or -OMe.
[0110] In some embodiments, the compound of formula A has a structure according to formula B: formula B, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein R1, R2, R9, m and n are as defined for the compounds having a structure according to formula A.
[0111] In some embodiments, the compound of formula A has a structure according to formula C: formula C, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein R1, R2, R9, m and n are as defined for the compounds having a structure according to formula A.
[0112] Examples of particular compounds of the present application include: and their pharmaceutically suitable salt, hydrate, solvate, polymorph, isomer or combination thereof.
[0113] Any embodiment described herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds, unless otherwise indicated. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H, 3H, UC, 13C, 14C, 15N, 18F, 31P, 32P, 35S, 36C1, 125I, respectively. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as¾, 13C, and 14C, are present. Such isotopically labeled compounds are useful in metabolic studies (preferably with 14C), reaction kinetic studies (with, for example 2H or ¾), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or labeled compound may be particularly preferred for PET or SPECT studies. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
[0114] Any of the individual embodiments recited herein may define formula V-a, A, B or C individually or be combined to produce a preferred embodiment of this invention.
General Synthetic Methodology
[0115] The compounds of this invention may be prepared in general by methods known to those skilled in the art. Schemes 1-9 below provide general synthetic routes for the preparation of compounds of formulae V-a, A, B and C. Other equivalent schemes, which will be readily apparent to the ordinary skilled organic chemist, may alternatively be used to synthesize various portions of the molecules as illustrated by the general schemes below.
Scheme 1. General synthesis of a compound of formula V-a, or a precursor to a compound of formula A or B wherein X, Y, Z, V and W form a 1,2,3-triazole ring, or a compound of formula B.
Scheme 2. General synthesis of a compound of formula V-a, or a precursor to a compound of formula A or B, wherein X, Y, Z, V and W form a phenoxy-substituted 1,2,3-triazole ring, or a compound of formula B. Scheme 3. General synthesis of compounds of formula V-a to allow for divergent functionalization on the triazolo-ring formed by X, Y, Z, V and W.
Scheme 4. General synthesis of a compound of formula V-a wherein X, Y, Z, V and W form an aminomethyl-substituted 1,2,3-triazole ring. Scheme 5. General synthesis of a compound of formula V-a wherein X, Y, Z, V and W form an aralkyl-substituted or heteroaralkyl substituted 1,2,3-triazole ring. Scheme 6. General synthesis of a compound of formula V-a, or precursors to compounds of formula A or C, wherein X, Y, Z, V and W form a substituted 1,2,4-triazole ring.
Scheme 7. General synthesis of a compound of formula V-a wherein X, Y, Z, V, and W form a methyl-substituted 1,2,3-triazole ring.
Scheme 8. General synthesis of a compound of formula V-a, wherein X, Y, Z, V, and W form a benzyl-substituted 1,2,3-triazole ring.
Scheme 9. General synthesis of a compound of formula V-a, A, B or C, where R3 of the compounds of formula V-a is an optionally substituted alkynyl group is illustrated in scheme 9. [0116] As would be recognized by skilled practitioners, compounds of formulae V-a, A,
B and C with variables other than those depicted above may be prepared by varying chemical reagents or the synthetic routes.
Pharmaceutical Compositions and Modes of Administration
[0117] The present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formulae V-a, A, B and C, or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof. [0118] The basic nitrogen-containing groups present in the compounds of the invention may be quatemized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates, such as dimethyl, diethyl, dibutyl and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides, such as benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained.
[0119] It will be appreciated that compounds and agents used in the compositions of this invention preferably should readily penetrate the blood-brain barrier when peripherally administered. Compounds which cannot penetrate the blood-brain barrier, however, can still be effectively administered directly into the central nervous system, e.g ., by an intraventricular or other neuro-compatible route.
[0120] In some embodiments of this invention, the a5-containing GABAA R positive allosteric modulator is formulated with a pharmaceutically acceptable carrier.
Pharmaceutically acceptable carriers that may be used in these compositions include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. In other embodiments, no carrier is used. For example, the a5-containing GABAA R agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator) can be administered alone or as a component of a pharmaceutical formulation (therapeutic composition). The a5-containing GABAA R agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator) may be formulated for administration in any convenient way for use in human medicine.
[0121] In some embodiments, the therapeutic methods of the invention include administering the composition of a compound or agent topically, systemically, or locally.
For example, therapeutic compositions of compounds or agents of the invention may be formulated for administration by, for example, injection (e.g, intravenously, subcutaneously, or intramuscularly), inhalation or insufflation (either through the mouth or the nose) or oral, buccal, sublingual, transdermal, nasal, or parenteral administration. The compositions of compounds or agents described herein may be formulated as part of an implant or device, or formulated for slow or extended release. When administered parenterally, the therapeutic composition of compounds or agents for use in this invention is preferably in a pyrogen-free, physiologically acceptable form. Techniques and formulations generally may be found in Remington’s Pharmaceutical Sciences, Meade Publishing Co., Easton, PA.
[0122] In certain embodiments, pharmaceutical compositions suitable for parenteral administration may comprise the a5-containing GABAA R positive allosteric modulator in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and non-aqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
[0123] A composition comprising a a5-containing GABAA R positive allosteric modulator may also contain adjuvants, such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.
[0124] In certain embodiments of the invention, compositions comprising a a5- containing GABAA R positive allosteric modulator can be administered orally, e.g. , in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and the like, each containing a predetermined amount of the a5- containing GABAA R positive allosteric modulator as an active ingredient.
[0125] In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules, and the like), one or more compositions comprising the a5-containing GABAA R positive allosteric modulator may be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
[0126] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the a5-containing GABAA R positive allosteric modulator, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol (ethanol), isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3- butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
[0127] Suspensions, in addition to the active compounds, may contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
[0128] As described herein, the compounds, agents, and compositions thereof may be administered for slow, controlled or extended release. The term “extended release” is widely recognized in the art of pharmaceutical sciences and is used herein to refer to a controlled release of an active compound or agent from a dosage form to an environment over (throughout or during) an extended period of time, e.g. greater than or equal to one hour. An extended release dosage form will release drug at substantially constant rate over an extended period of time or a substantially constant amount of drug will be released incrementally over an extended period of time. The term “extended release” used herein includes the terms “controlled release,” “prolonged release,” “sustained release,” “delayed release,” or “slow release” as these terms are used in the pharmaceutical sciences. In some embodiments, the extended release dosage is administered in the form of a patch or a pump.
[0129] A person of ordinary skill in the art, such as a physician, is readily able to determine the required amount of a5-containing GABAA R positive allosteric modulator (s) to treat the subject using the compositions and methods of the invention. It is understood that the dosage regimen will be determined for an individual, taking into consideration, for example, various factors that modify the action of a5-containing GABAA R positive allosteric modulator, the severity or stage of the disease, route of administration, and characteristics unique to the individual, such as age, weight, size, and extent of cognitive impairment.
[0130] It is well-known in the art that normalization to body surface area is an appropriate method for extrapolating doses between species. To calculate the human equivalent dose (HED) from a dosage used in the treatment of age-dependent cognitive impairment in rats, the formula HED (mg/kg) = rat dose (mg/kg) x 0.16 may be employ ed(see Estimating the Safe Starting Dose in Clinical Trials for Therapeutics in Adult Healthy Volunteers, December 2002, Center for Biologies Evaluation and Research). For example, using that formula, a dosage of 10 mg/kg in rats is equivalent to 1.6 mg/kg in humans. This conversion is based on a more general formula HED = animal dose in mg/kg x (animal weight in kg/human weight in kg) 0 3.
[0131] In certain embodiments of the invention, the dose of the a5-containing GABAA R positive allosteric modulator is between 0.0001 and 100 mg/kg/day (which, given a typical human subject of 70 kg, is between 0.007 and 7000 mg/day).
[0132] In certain embodiments of the invention, the interval of administration is once every 12 or 24 hours. Administration at less frequent intervals, such as once every 6 hours, may also be used.
[0133] If administered by an implant, a device or a slow or extended release formulation, the a5-containing GABAA R positive allosteric modulator can be administered one time, or one or more times periodically throughout the lifetime of the patient as necessary.
Other administration intervals intermediate to or shorter than these dosage intervals for clinical applications may also be used and may be determined by one skilled in the art following the methods of this invention.
[0134] Desired time of administration can be determined by routine experimentation by one skilled in the art. For example, the a5-containing GABAA R positive allosteric modulator may be administered for a period of 1-4 weeks, 1-3 months, 3-6 months, 6-12 months, 1-2 years, or more, up to the lifetime of the patient.
[0135] In addition to a5-containing GABAA R positive allosteric modulator, the compositions of this invention can also include other therapeutically useful agents. These other therapeutically useful agents may be administered in a single formulation, simultaneously or sequentially with the a5-containing GABAA R positive allosteric modulator according to the methods of the invention.
[0136] It will be understood by one of ordinary skill in the art that the compositions described herein may be adapted and modified as is appropriate for the application being addressed and that the compositions described herein may be employed in other suitable applications. For example, the compositions of this application may further comprise a second therapeutic agent. Such other additions and modifications will not depart from the scope hereof. Pharmaceutical Compositions with Antipsychotics
[0137] The compounds or the compositions of this application may be used in combination with an antipsychotic in treating cognitive impairment associated with schizophrenia or bipolar disorder in a subject having or at risk of said schizophrenia or bipolar disorder ( e.g ., mania). The antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof that is useful in the methods and compositions of this invention include both typical and atypical antipsychotics. In some embodiments, the compounds or the compositions of the present invention may be used to treat one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia. In some embodiments, the compounds or the compositions of the present invention may be used to treat one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments of this invention, the compounds or the compositions of this invention prevent or slow the progression of cognitive impairment of schizophrenia or bipolar disorder (in particular, mania) in said subject.
[0138] In some embodiments, the antipsychotics suitable for use in the present invention are selected from atypical antipsychotics. Such atypical antipsychotics include, but are not limited to, those disclosed in, for example, U.S. Patents 4,734,416; 5,006,528; 4,145,434; 5,763,476; 3,539,573; 5,229,382; 5,532,372; 4,879,288; 4,804,663; 4,710,500; 4,831,031; and 5,312,925, and EP Patents EP402644 and EP368388, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
[0139] In some embodiments, atypical antipsychotics suitable for use in the present invention include, but are not limited to, aripiprazole, asenapine, clozapine, iloperidone, olanzapine, lurasidone, paliperidone, quetiapine, risperidone and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof. In some embodiments, the antipsychotic suitable for use herein is selected from aripiprazole (Bristol-Myers Squibb), olanzapine (Lilly) and ziprasidone (Pfizer), and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
[0140] In some embodiments, the antipsychotics suitable for use in the present invention are typical antipsychotics, including, but not limited to, acepromazine, benperidol, bromazepam, bromperidol, chlorpromazine, chlorprothixene, clotiapine, cyamemazine, diazepam, dixyrazine, droperidol, flupentixol, fluphenazine, fluspirilene, haloperidol, heptaminol, isopropamide iodide, levomepromazine, levosulpiride, loxapine, melperone, mesoridazine, molindone, oxypertine, oxyprothepine, penfluridol, perazine, periciazine, perphenazine, pimozide, pipamperone, pipotiazine, prochlorperazine, promazine, promethazine, prothipendyl, pyridoxine, sulpiride, sultopride, tetrabenazine, thioproperazine, thioridazine, tiapride, tiotixene, trifluoperazine, triflupromazine, trihexyphenidyl, and zuclopenthixol, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
[0141] In some embodiments of the present invention, the antipsychotic or a pharmaceutically acceptable salt, hydrate, solvate or polymorph thereof may be selected from compounds that are dopaminergic agents (such as dopamine D1 receptor antagonists or agonists, dopamine D2 receptor antagonists or partial agonists, dopamine D3 receptor antagonists or partial agonists, dopamine D4 receptor antagonists), glutamatergic agents, N-methyl-D-aspartate (NMD A) receptor positive allosteric modulators, glycine reuptake inhibitors, glutamate reuptake inhibitor, metabotropic glutamate receptors (mGluRs) agonists or positive allosteric modulators (PAMs) ( e.g ., mGluR2/3 agonists or PAMs), glutamate receptor glur5 positive allosteric modulators (PAMs), Ml muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs), histamine H3 receptor antagonists, a-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)/kainate receptor antagonists, ampakines (CX-516), glutathione prodrugs, noradrenergic agents (such as alpha-2 adrenergic receptor agonists or antagonists and catechol-O-methyl transferase (COMT) inhibitors), serotonin receptor modulators (such as 5-HT2A receptor antagonists, 5-HTIA receptor partial agonists, 5-HT2c agonists, and 5-HT6 antagonists, serotonin 2C agonists), cholinergic agents (such as alpha-7 nicotinic receptor agonists or PAMs, alpha4-beta2 nicotinic receptor agonists, allosteric modulators of nicotinic receptors and acetylcholinesterase inhibitors, muscarinic receptor agonists and antagonists), cannabinoid CB1 antagonists, neurokinin 3 antagonists, neurotensin agonists, monoamine oxidase (MAO) B inhibitors, PDE10 inhibitors, neuronal nitric oxide synthase (nNOS) inhibitors, neurosteroids, and neurotrophic factors.
[0142] In some embodiments, an a5-containing GABAA receptor positive allosteric modulator as described herein and an antipsychotic as described herein, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs, are administered simultaneously, or sequentially, or in a single formulation, or in separate formulations packaged together. In other embodiments, the a5-containing GABAA receptor positive allosteric modulator and the antipsychotic, or their pharmaceutically acceptable salts, hydrates, solvates or polymorphs, are administered via different routes. As used herein, “combination” includes administration by any of these formulations or routes of administration.
Pharmaceutical Compositions with Memantine
[0143] The compounds or the compositions of this application may be used in combination with memantine or a derivative or an analog thereof in treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI, Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia or bipolar disorder, amyotrophic lateral sclerosis (ALS) and cancer-therapy- related cognitive impairment.
[0144] Memantine, chemically also known as 3,5-dimethyladamantan-l-amine or 3,5- dimethyltricyclo[3.3.1.13,7]decan-l-amine, is an uncompetitive N-methyl-D-aspartate (NMD A) receptor antagonist with moderate affinity. The proprietary names for memantine include: Axura® and Akatinol® (Merz), Namenda® (Forest Laboratories), Ebixa® and Abixa® (Lundbeck), and Memox® (Unipharm). Memantine is currently available in the U.S. and in over 42 countries worldwide. It is approved for the treatment of moderate to severe Alzheimer's disease (AD) in the United States at a dose of up to 28 mg/day. Memantine and some of its derivatives and analogs that are useful in the present invention are disclosed in U.S. Patents Nos. 3,391,142; 4,122,193; 4,273,774; and 5,061,703, all of which are hereby incorporated by reference. Other memantine derivatives or analogs that are useful in the present invention include, but are not limited to, those compounds disclosed in U.S. Patent Application Publication US20040087658, US20050113458, US20060205822, US20090081259, US20090124659, and US20100227852; EP Patent Application Publication EP2260839A2; EP Patent EP1682109B1; and PCT Application Publication W02005079779, all of which are incorporated herein by reference. Memantine, as used in the present invention, includes memantine and its derivatives and analogs, as well as hydrates, polymorphs, prodrugs, salts, and solvates thereof. Memantine, as used herein, also includes a composition comprising memantine or a derivative or an analog or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, or prodrug thereof, wherein the composition optionally further comprises at least one additional therapeutic agent (such as a therapeutic agent useful for treating a CNS disorder or cognitive impairments associated thereof). In some embodiments, the memantine composition suitable for use in the present invention comprises memantine and a second therapeutic agent that is donepezil (under the trade name Aricept).
[0145] In other embodiments of the invention, the a5-containing GABAA receptor positive allosteric modulator and memantine (or the memantine derivative/analog), or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, or prodrugs are administered simultaneously, or sequentially, or in a single formulation or in separate formulations packaged together. In other embodiments, the a5-containing GABAA receptor positive allosteric modulator and memantine (or the memantine derivative/analog), or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, or prodrugs are administered via different routes. As used herein, "combination" includes administration by any of these formulations or routes of administration.
Pharmaceutical Compositions with Acetylcholine Esterase Inhibitors (AChE-Is)
[0146] The compounds or the compositions of this application may be used in combination with an acetylcholine esterase inhibitor in treating cognitive impairment associated with central nervous system (CNS) disorders in a subject in need or at risk thereof, including, without limitation, subjects having or at risk for age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI, Age-Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease(AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia or bipolar disorder, amyotrophic lateral sclerosis (ALS) and cancer-therapy-related cognitive impairment.
[0147] AChE-Is known to a person of ordinary skill in the art may belong to the subcategories of (i) reversible non-competitive inhibitors or reversible competitive inhibitors, (ii) irreversible, and/or (iii) quasi-irreversible inhibitors.
[0148] In certain embodiment, AChE-Is useful in the present invention include those described in PCT applications W02014039920 and W02002032412; EP patents Nos. 468187; 481429-A; and U.S. Patents Nos. 4,816,456; 4,895,841; 5,041,455; 5,106,856; 5,602,176; 6,677,330; 7,340,299; 7,635,709; 8,058,268; 8,741,808; and 8,853,219, all of which are incorporated herein by reference.
[0149] In certain embodiment, typical AChE-Is that may be used in accordance with this invention include, but are not limited to, ungeremine, ladostigil, demecarium, echothiophate (Phospholine), edrophonium (Tensilon), tacrine (Cognex), Pralidoxime (2- PAM), pyridostigmine (Mestinon), physostigmine (serine, Antilirium), abmenonium (Mytelase), galantamine (Reminyl, Razadyne), rivastigmine (Exelon, SZD-ENA-713), Huperzine A, Icopezil, neostigmine (Prostigmin, Vagostigmin), Aricept (Donepezil, E2020), Lactucopicrin, monoamine acridines and their derivatives, piperidine and piperazine derivatives, N-benzyl-piperidine derivatives, piperidinyl-alkanoyl heterocyclic compounds , 4-(l -benzyl :piperidyl)-substituted fused quinoline derivatives and cyclic amide derivatives. Other typical AChE-Is include carbamates and organophosphonate compounds such as Metrifonate (Trichlorfon). Benzazepinols such as galantamine are also useful AChE-Is. In some embodiment, AChE-Is suitable for use in combination with the compounds and compositions of this application include: Donepezil (aricept), Galantamine (razadyne), or Rivastigmine (exelon).
[0150] In other embodiments of the invention, the a5-containing GABAA receptor positive allosteric modulator and the AChE-I, or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, or prodrugs are administered simultaneously, or sequentially, or in a single formulation or in separate formulations packaged together. In other embodiments, the a5-containing GABAA receptor positive allosteric modulator and the AChE-I, or their pharmaceutically acceptable salts, hydrates, solvates, polymorphs, or prodrugs are administered via different routes. As used herein, "combination" includes administration by any of these formulations or routes of administration.
[0151] In some embodiments, the compounds and compositions described herein are for use as a medicament. In some embodiments, the compounds and compositions of the present invention are for use in treating cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
[0152] In some embodiments, this application provides the use of a compound or composition described herein in the preparation of a medicament for the treatment of cognitive impairment associated with a CNS disorder in a subject in need of treatment or at risk of said cognitive impairment. In some embodiments, the CNS disorder with cognitive impairment includes, without limitation, age-related cognitive impairment, Mild Cognitive Impairment (MCI), amnestic MCI (aMCI), Age- Associated Memory Impairment (AAMI), Age Related Cognitive Decline (ARCD), dementia, Alzheimer’s Disease (AD), prodromal AD, post-traumatic stress disorder (PTSD), schizophrenia, bipolar disorder, amyotrophic lateral sclerosis (ALS), cancer-therapy-related cognitive impairment, mental retardation, Parkinson’s disease (PD), autism spectrum disorders, fragile X disorder, Rett syndrome, compulsive behavior, and substance addiction.
Methods of assessing cognitive impairment
[0153] Animal models serve as an important resource for developing and evaluating treatments for cognitive impairment associated with CNS disorders. Features that characterize cognitive impairment in animal models typically extend to cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans. The extent of cognitive impairment in an animal model for a CNS disorder, and the efficacy of a method of treatment for said CNS disorder may be tested and confirmed with the use of a variety of cognitive tests.
[0154] A Radial Arm Maze (RAM) behavioral task is one example of a cognitive test, specifically testing spacial memory (Chappell et al. Neuropharmacology 37: 481-487, 1998). The RAM apparatus consists of, e.g. , eight equidistantly spaced arms. A maze arm projects from each facet of a center platform. A food well is located at the distal end of each arm. Food is used as a reward. Blocks can be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus may also be provided. After habituation and training phases, spatial memory of the subjects may be tested in the RAM under control or test compound-treated conditions. As a part of the test, subjects are pretreated before trials with a vehicle control or one of a range of dosages of the test compound. At the beginning of each trial, a subset of the arms of the eight-arm maze is blocked. Subjects are allowed to obtain food on the unblocked arms to which access is permitted during this initial “information phase” of the trial. Subjects are then removed from the maze for a delay period, e.g ., a 60 second delay, a 15 minute delay, a one-hour delay, a two-hour delay, a six hour delay, a 24 hour delay, or longer) between the information phase and the subsequent “retention test,” during which the barriers on the maze are removed, thus allowing access to all eight arms. After the delay period, subjects are placed back onto the center platform (with the barriers to the previously blocked arms removed) and allowed to obtain the remaining food rewards during this retention test phase of the trial. The identity and configuration of the blocked arms vary across trials. The number of “errors” the subjects make during the retention test phase is tracked. An error occurs in the trial if the subjects entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if it re-visits an arm in the post-delay session that had already been visited. A fewer number of errors would indicate better spatial memory. The number of errors made by the test subject, under various test compound treatment regimes, can then be compared for efficacy of the test compound in treating cognitive impairment associated with CNS disorders.
[0155] Another cognitive test that may be used to assess the effects of a test compound on the cognitive impairment of a CNS disorder model animal is the Morris water maze. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampal-dependent (de Hoz et al., Eur. J. Neurosci ., 22:745-54, 2005; Steele and Morris, Hippocampus 9: 118-36, 1999). The subject is trained to locate a submerged escape platform hidden underneath the surface of the pool. During the training trial, a subject is released in the maze (pool) from random starting positions around the perimeter of the pool. The starting position varies from trial to trial. If the subject does not locate the escape platform within a set time, the experimenter guides and places the subject on the platform to “teach” the location of the platform. After a delay period following the last training trial, a retention test in the absence of the escape platform is given to assess spatial memory. The subject’s level of preference for the location of the (now absent) escape platform, as measured by, e.g ., the time spent in that location or the number of crossings of that location made by the mouse, indicates better spatial memory, i.e., treatment of cognitive impairment. The preference for the location of the escape platform under different treatment conditions, can then be compared for efficacy of the test compound in treating cognitive impairment associated with CNS disorders.
[0156] There are various tests known in the art for assessing cognitive function in humans, for example and without limitation, the clinical global impression of change scale (CIBIC-plus scale); the Mini Mental State Exam (MMSE); the Neuropsychiatric Inventory (NPI); the Clinical Dementia Rating Scale (CDR); the Cambridge Neuropsychological Test Automated Battery (CANTAB); the Sandoz Clinical Assessment-Geriatric (SC AG), the Buschke Selective Reminding Test (Buschke and Fuld, 1974); the Verbal Paired Associates subtest; the Logical Memory subtest; the Visual Reproduction subtest of the Wechsler Memory Scale-Revised (WMS-R) (Wechsler, 1997); the Benton Visual Retention Test, or MATRICS consensus neuropsychological test battery which includes tests of working memory, speed of processing, attention, verbal learning, visual learning, reasoning and problem solving and social cognition. See Folstein et ah, J Psychiatric Res 12: 189-98, (1975); Robbins et al., Dementia 5: 266-81, (1994); Rey, L'examen clinique en psychologic, (1964); Kluger et al., J Geriatr Psychiatry Neurol 12:168-79, (1999); Marquis et al., 2002 and Masur et al., 1994. Also see Buchanan, R.W., Keefe, R.S.E., Umbricht, D., Green, M.F., Laughren, T., and Marder, S.R. (2011) The FDA-NIMH- MATRICS guidelines for clinical trial design of cognitive-enhancing drugs: what do we know 5 years later? Schizophr. Bull. 37, 1209-1217. Another example of a cognitive test in humans is the explicit 3-alternative forced choice task. In this test, subjects are presented with color photographs of common objects consisting of a mix of three types of image pairs: similar pairs, identical pairs and unrelated foils. The second of the pair of similar objects is referred to as the "lure". These image pairs are fully randomized and presented individually as a series of images. Subjects are instructed to make a judgment as to whether the objects seen are new, old or similar. A “similar” response to the presentation of a lure stimulus indicates successful memory retrieval by the subject. By contrast, calling the lure stimulus “old” or “new” indicates that correct memory retrieval did not occur.
[0157] In addition to assessing cognitive performance, the progression of age-related cognitive impairment and dementia, as well as the conversion of age-related cognitive impairment into dementia, may be monitored by assessing surrogate changes in the brain of the subject. Surrogate changes include, without limitation, changes in regional brain volumes, perforant path degradation, and changes seen in brain function through resting state fMRI (R-fMRI) and fluorodeoxyglucose positron emission tomography (FDG-PET). Examples of regional brain volumes useful in monitoring the progression of age-related cognitive impairment and dementia include reduction of hippocampal volume and reduction in volume or thickness of entorhinal cortex. These volumes may be measured in a subject by, for example, MRI. Aisen et al., Alzheimer’s & Dementia 6:239-246 (2010). Perforant path degradation has been shown to be linked to age, as well as reduced cognitive function. For example, older adults with more perforant path degradation tend to perform worse in hippocampus-dependent memory tests. Perforant path degradation may be monitored in subjects through ultrahigh-resolution diffusion tensor imaging (DTI). Yassa et al., PNAS 107:12687-12691 (2010). Resting-state fMRI (R-fMRI) involves imaging the brain during rest, and recording large-amplitude spontaneous low-frequency (<0.1 Hz) fluctuations in the fMRI signal that are temporally correlated across functionally related areas. Seed-based functional connectivity, independent component analyses, and/or frequency-domain analyses of the signals are used to reveal functional connectivity between brain areas, particularly those areas whose connectivity increase or decrease with age, as well as the extent of cognitive impairment and/or dementia. FDG-PET uses the uptake of FDG as a measure of regional metabolic activity in the brain. Decline of FDG uptake in regions such as the posterior cingulated cortex, temporoparietal cortex, and prefrontal association cortex has been shown to relate to the extent of cognitive decline and dementia. Aisen et al., Alzheimer’s & Dementia 6:239-246 (2010), Herholz et al., Neuroimage 17:302-316 (2002).
Age-Related Cognitive Impairment
[0158] The invention provides methods and compositions for treating age-related cognitive impairment or the risk thereof using a a5 -containing GABAA receptor positive allosteric modulator (i.e., a compound of the invention), such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of age-related cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with age-related cognitive impairment. In certain embodiments, treatment of age-related cognitive impairment comprises slowing the conversion of age-related cognitive impairment (including, but not limited to MCI, ARCD and AAMI) into dementia ( e.g ., AD). The methods and compositions may be used for human patients in clinical applications in the treating age-related cognitive impairment in conditions such as MCI, ARCD and AAMI or for the risk thereof. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with age-related cognitive impairment, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0159] In some embodiments, a subject to be treated by the methods and compositions of this invention exhibits age-related cognitive impairment or is at risk of such impairment. In some embodiments, the age-related cognitive impairment includes, without limitation, Age-Associated Memory Impairment (AAMI), Mild Cognitive Impairment (MCI) and Age-related Cognitive Decline (ARCD).
[0160] Animal models serve as an important resource for developing and evaluating treatments for such age-related cognitive impairments. Features that characterize age- related cognitive impairment in animal models typically extend to age-related cognitive impairment in humans. Efficacy in such animal models is, thus, expected to be predictive of efficacy in humans.
[0161] Various animal models of age-related cognitive impairment are known in the art. For example, extensive behavioral characterization has identified a naturally occurring form of cognitive impairment in an outbred strain of aged Long-Evans rats (Charles River Laboratories; Gallagher et al., Behav. Neurosci. 107:618-626, (1993)). In a behavioral assessment with the Morris Water Maze (MWM), rats learn and remember the location of an escape platform guided by a configuration of spatial cues surrounding the maze. The cognitive basis of performance is tested in probe trials using measures of the animal’s spatial bias in searching for the location of the escape platform. Aged rats in the study population have no difficulty swimming to a visible platform, but an age-dependent impairment is detected when the platform is camouflaged, requiring the use of spatial information. Performance for individual aged rats in the outbred Long-Evans strain varies greatly. For example, a proportion of those rats perform on a par with young adults. However, approximately 40-50% fall outside the range of young performance. This variability among aged rats reflects reliable individual differences. Thus, within the aged population some animals are cognitively impaired and designated aged-impaired (AI) and other animals are not impaired and are designated aged-unimpaired (AU). See, e.g., Colombo eta/., Proc. Natl. Acad. Sci. 94: 14195-14199, (1997); Gallagher and Burwell, Neurobiol. Aging 10: 691-708, (1989); Gallagher et al. Behav. Neurosci. 107:618-626, (1993); Rapp and Gallagher, Proc. Natl. Acad. Sci. 93: 9926-9930, (1996); Nicolle et al, Neuroscience 74: 741-756, (1996); Nicolle et al, J. Neurosci. 19: 9604-9610, (1999); International Patent Publication W02007/019312 and International Patent Publication WO 2004/048551. Such an animal model of age-related cognitive impairment may be used to assay the effectiveness of the methods and compositions this invention in treating age- related cognitive impairment.
[0162] The efficacy of the methods and compositions of this invention in treating age- related cognitive impairment may be assessed using a variety of cognitive tests, including the Morris water maze and the radial arm maze, as discussed herein.
Dementia
[0163] The invention also provides methods and compositions for treating dementia using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of dementia. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with dementia. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with dementia, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In certain embodiments, the dementia is Alzheimer’s disease (AD), vascular dementia, dementia with Lewy bodies, or frontotemporal dementia. The methods and compositions may be used for human patients in clinical applications in treating dementia. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
[0164] Animal models serve as an important resource for developing and evaluating treatments for dementia. Features that characterize dementia in animal models typically extend to dementia in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of dementia are known in the art, such as the PDAPP, Tg2576, APP23, TgCRND8, J20, hPS2 Tg, and APP + PS1 transgenic mice. Sankaranarayanan, Curr. Top. Medicinal Chem. 6: 609-627, 2006; Kobayashi et al. Genes Brain Behav. 4: 173-196. 2005; Ashe and Zahns, Neuron. 66: 631 - 45, 2010. Such animal models of dementia may be used to assay the effectiveness of the methods and compositions of this invention of the invention in treating dementia.
[0165] The efficacy of the methods and compositions of this invention in treating dementia, or cognitive impairment associated with dementia, may be assessed in animals models of dementia, as well as human subjects with dementia, using a variety of cognitive tests known in the art, as discussed herein.
Post-Traumatic Stress Disorder
[0166] The invention also provides methods and compositions for treating post-traumatic stress disorder (PTSD) using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of PTSD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with PTSD. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with PTSD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating PTSD. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
[0167] Patients with PTSD (and, to a lesser degree, trauma-exposed patients without PTSD) have smaller hippocampal volumes (Woon etal. , Prog. Neuro-Psychopharm. & Biological Psych. 34, 1181-1188; Wang et al, Arch. Gen. Psychiatry 67 : 296-303 , 2010). PTSD is also associated with impaired cognitive performance. Older individuals with PTSD have greater declines in cognitive performance relative to control patients (Yehuda etal. , Bio. Psych. 60: 714-721, 2006) and have a greater likelihood of developing dementia (Yaffe etal, Arch. Gen. Psych. 678: 608-613, 2010).
[0168] Animal models serve as an important resource for developing and evaluating treatments for PTSD. Features that characterize PTSD in animal models typically extend to PTSD in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of PTSD are known in the art.
[0169] One rat model of PTSD is Time-dependent sensitization (TDS). TDS involves exposure of the animal to a severely stressful event followed by a situational reminder of the prior stress. The following is an example of TDS. Rats are placed in a restrainer, then placed in a swim tank and made to swim for a period of time, e.g. , 20 min. Following this, each rat is then immediately exposed to a gaseous anesthetic until loss of consciousness, and finally dried. The animals are left undisturbed for a number of days, e.g. , one week. The rats are then exposed to a “restress” session consisting of an initial stressor, e.g. , a swimming session in the swim tank (Liberzon et al. , Psychoneuroendocrinology 22: 443- 453, 1997; Harvery etal. , Psychopharmacology 175:494-502, 2004). TDS results in an enhancement of the acoustic startle response (ASR) in the rat, which is comparable to the exaggerated acoustic startle that is a prominent symptom of PTSD (Khan and Liberzon, Psychopharmacology 172: 225-229, 2004). Such animal models of PTSD may be used to assay the effectiveness of the methods and compositions of this invention of the invention in treating PTSD. [0170] The efficacy of the methods and compositions of this invention in treating PTSD, or cognitive impairment associated with PTSD, may also be assessed in animals models of PTSD, as well as human subjects with PTSD, using a variety of cognitive tests known in the art, as discussed herein.
Schizophrenia and Bipolar Disorder
[0171] The invention additionally provides methods and compositions for treating schizophrenia or bipolar disorder (in particular, mania) using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of schizophrenia or bipolar disorder (in particular, mania). Schizophrenia is characterized by a wide spectrum of psychopathology, including positive symptoms such as aberrant or distorted mental representations ( e.g ., hallucinations, delusions), or dopamine dysregulati on-associated symptoms (e.g., hyperdopaminergic responses, hyperdopaminergic behavioral responses, dopaminergic hyperactivity, or hyperlocomotor activity, or psychosis), negative symptoms characterized by diminution of motivation and adaptive goal-directed action (e.g, anhedonia, affective flattening, avolition), and cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more positive and/or negative symptoms, as well as cognitive impairment, associated with schizophrenia. Further, there are a number of other psychiatric diseases such as schizotypical and schizoaffective disorder, other acute and chronic psychoses and bipolar disorder (in particular, mania), which have an overlapping symptomatology with schizophrenia. In some embodiments, treatment comprises alleviation, amelioration or slowing the progression of one or more symptoms, as well as cognitive impairment, associated with bipolar disorder (in particular, mania). In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with schizophrenia or bipolar disorder, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating schizophrenia or bipolar disorder (in particular, mania). The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
[0172] Cognitive impairments are associated with schizophrenia. They precede the onset of psychosis and are present in non-affected relatives. The cognitive impairments associated with schizophrenia constitute a good predictor for functional outcome and are a core feature of the disorder. Cognitive features in schizophrenia reflect dysfunction in frontal cortical and hippocampal circuits. Patients with schizophrenia also present hippocampal pathologies such as reductions in hippocampal volume, reductions in neuronal size and dysfunctional hyperactivity. An imbalance in excitation and inhibition in these brain regions has also been documented in schizophrenic patients suggesting that drugs targeting inhibitory mechanisms could be therapeutic. See , e.g. , Guidotti el al ., Psychopharmacology 180: 191-205, 2005; Zierhut , Psych. Res. Neuroimag. 183:187-194, 2010; Wood et al. , Neuroimage 52:62-63, 2010; Vinkers et al. , Expert Opin. Investig. Drugs 19:1217-1233, 2009; Young et al. , Pharmacol. Ther. 122:150-202, 2009.
[0173] Animal models serve as an important resource for developing and evaluating treatments for schizophrenia. Features that characterize schizophrenia in animal models typically extend to schizophrenia in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of schizophrenia are known in the art.
[0174] One animal model of schizophrenia is protracted treatment with methionine. Methionine-treated mice exhibit deficient expression of GAD67 in frontal cortex and hippocampus, similar to those reported in the brain of postmortem schizophrenia patients. They also exhibit prepulse inhibition of startle and social interaction deficits (Tremonlizzo et al, PNAS, 99: 17095-17100, 2002). Another animal model of schizophrenia is methylaoxymethanol acetate (MAM)-treatment in rats. Pregnant female rats are administered MAM (20 mg/kg, intraperitoneal) on gestational day 17. MAM-treatment recapitulate a pathodevelopmental process to schizophrenia-like phenotypes in the offspring, including anatomical changes, behavioral deficits and altered neuronal information processing. More specifically, MAM-treated rats display a decreased density of parvalbumin-positive GABAergic interneurons in portions of the prefrontal cortex and hippocampus. In behavioral tests, MAM-treated rats display reduced latent inhibition. Latent inhibition is a behavioral phenomenon where there is reduced learning about a stimulus to which there has been prior exposure with any consequence. This tendency to disregard previously benign stimuli, and reduce the formation of association with such stimuli is believed to prevent sensory overload. Low latent inhibition is indicative of psychosis. Latent inhibition may be tested in rats in the following manner. Rats are divided into two groups. One group is pre-exposed to a tone over multiple trials. The other group has no tone presentation. Both groups are then exposed to an auditory fear conditioning procedure, in which the same tone is presented concurrently with a noxious stimulus, e.g. an electric shock to the foot. Subsequently, both groups are presented with the tone, and the rats’ change in locomotor activity during tone presentation is monitored. After the fear conditioning the rats respond to the tone presentation by strongly reducing locomotor activity. However, the group that has been exposed to the tone before the conditioning period displays robust latent inhibition: the suppression of locomotor activity in response to tone presentation is reduced. MAM-treated rats, by contrast show impaired latent inhibition. That is, exposure to the tone previous to the fear conditioning procedure has no significant effect in suppressing the fear conditioning (see Lodge el al ., J. Neurosci., 29:2344-2354, 2009) Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
[0175] MAM-treated rats display a significantly enhanced locomotor response (or aberrant locomotor activity) to low dose D-amphetamine administration. The MAM- treated rats also display a significantly greater number of spontaneously firing ventral tegmental dopamine (DA) neurons. These results are believed to be a consequence of excessive hippocampal activity because in MAM-treated rats, the ventral hippocampus (vHipp) inactivation (e.g, by intra-vHipp administration of a sodium channel blocker, tetrodotoxin (TTX), to MAM rats) completely reversed the elevated DA neuron population activity and also normalized the augmented amphetamine-induced locomotor behavior. The correlation of hippocampal dysfunction and the hyper-responsivity of the DA system is believed to underlie the augmented response to amphetamine in MAM- treated animals and psychosis in schizophrenia patients. See Lodge D. J. et al. Neurobiology of Disease (2007), 27(42), 11424-11430. The use of MAM-treated rats in the above study may be suitable for use to assay the effectiveness of the methods and compositions of the present invention in treating schizophrenia or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the central hippocampus (vHipp) regulation, on the elevated DA neuron population activity and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
[0176] In MAM-treated rats, hippocampal (HPC) dysfunction leads to dopamine system hyperactivity. A benzodiazepine-positive allosteric modulator (PAM), selective for the a5 subunit of the GABAA receptor, SH-053-2’F-R-CH is tested for its effects on the output of the hippocampal (HPC). The effect of SH-053-2T-R-CH3 on the hyperactive locomotor response to amphetamine in MAM-treated animals is also examined. The a5GABAAR PAM reduces the number of spontaneously active DA neurons in the ventral tegmental area (VTA) of MAM rats to levels observed in saline-treated rats (control group), both when administered systemically and when directly infused into the ventral HPC. Moreover, HPC neurons in both saline-treated and MAM-treated animals show diminished cortical-evoked responses following the a5GABAAR PAM treatment. In addition, the increased locomotor response to amphetamine observed in MAM-treated rats is reduced following the OI5GABAAR PAM treatment. See Gill K. M et al. Neuropsychopharmacology (2011), 1-9. The use of MAM-treated rats in the above study may be suitable for use in the present invention to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania). For example, the methods and compositions of this invention maybe evaluated, using MAM-treated animals, for their effects on the output of the hippocampal (HPC) and on the hyperactive locomotor response to amphetamine in the MAM-treated animals.
[0177] Administration of MAM to pregnant rats on embryonic day 15 (E15) severely impairs spatial memory or the ability to learn the spatial location of four items on an eight- arm radial maze in the offspring. In addition, embryonic day 17 (El 7) MAM-treated rats are able to reach the level of performance of control rats at the initial stages of training, but are unable to process and retrieve spatial information when a 30-min delay is interposed, indicating a significant impairment in working memory. See Gourevitch R. et al. (2004). Behav. Pharmacol , 15, 287-292. Such animal models of schizophrenia may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania). [0178] Apomorphine-induced climbing (AIC) and stereotype (AIS) in mice is another animal model useful in this invention. Agents are administered to mice at a desired dose level ( e.g ., via intraperitoneal administration). Subsequently, e.g. , thirty minutes later, experimental mice are challenges with apomorphine (e.g., with 1 mg/kg sc). Five minutes after the apomorphine injection, the sniffmg-licking-gnawing syndrome (stereotyped behavior) and climbing behavior induced by apomorphine are scored and recorded for each animal. Readings can be repeated every 5 min during a 30-min test session. Scores for each animal are totaled over the 30-min test session for each syndrome (stereotyped behavior and climbing). If an effect reached at least of 50% inhibition, and ID50 value (95% confidence interval) is calculated using a nonlinear least squares calculation with inverse prediction. Mean climbing and stereotype scores can be expressed as a percent of control values observed in vehicle treated (e.g, saline-treated) mice that receive apomorphine. See Grauer S. M. et al. Psychopharmacology (2009) 204, 37-48. This mouse model may be used to assay the effectiveness of the methods and compositions of the invention in treating schizophrenia or bipolar disorder (in particular, mania).
[0179] In another well-established preclinical model of schizophrenia, rats exposed chronically to ketamine, an uncompetitive N-methyl-D-aspartate (NMD A) receptor antagonist, produces positive and negative psychotic symptoms and cognitive impairment. Long-Evans male rats are injected intraperitoneally with ketamine (30 mg/kg, twice a day) for two weeks during adolescence (2 month-old). Rats are behaviorally tested when they reach adulthood (approximately 4-5 month-old) for the behavioral symptoms to ketamine exposure and for the efficacy of treatment to alleviate those symptoms. See, e.g, Enomoto et al. Progress in Neuro-Psychopharmacology & Biological Psychiatry 33 (2009) 668- 675.
[0180] The efficacy of the methods and compositions of this invention in treating schizophrenia or cognitive impairment associated therewith may also be assessed in animal models of schizophrenia or bipolar disorder (in particular, mania), as well as human subjects with schizophrenia, using a variety of cognitive tests known in the art, as discussed herein. Amyotrophic Lateral Sclerosis (ALS)
[0181] The invention additionally provides methods and compositions for treating ALS using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of ALS. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with ALS. In certain embodiments, the symptom to be treated is cognitive impairment. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with ALS, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating ALS. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
[0182] In addition to the degeneration of motor neurons, ALS is characterized by neuronal degeneration in the entorhinal cortex and hippocampus, memory deficits, and neuronal hyperexcitability in different brain areas such as the cortex.
[0183] The efficacy of the methods and compositions of this invention in treating ALS, or cognitive impairment associated with ALS, may also be assessed in animal models of ALS, as well as human subjects with ALS, using a variety of cognitive tests known in the art, as discussed herein.
Cancer therapy-related cognitive impairment
[0184] The invention additionally provides methods and compositions for treating cancer therapy-related cognitive impairment using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression, of cancer therapy-related cognitive impairment. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with cancer therapy-related cognitive impairment. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with cancer therapy-related cognitive impairment, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. The methods and compositions may be used for human patients in clinical applications in treating cancer therapy-related cognitive impairment. The dose of the composition and dosage interval for the method is, as described herein, one that is safe and efficacious in those applications.
[0185] Therapies that are used in cancer treatment, including chemotherapy, radiation, or combinations thereof, can cause cognitive impairment in patients, in such functions as memory, learning and attention. Cytotoxicity and other adverse side-effects on the brain of cancer therapies are the basis for this form of cognitive impairment, which can persist for decades. (Dietrich et al. , Oncologist 13:1285-95, 2008; Soussain et al. , Lancet 374:1639-51, 2009).
[0186] Cognitive impairment following cancer therapies reflects dysfunction in frontal cortical and hippocampal circuits that are essential for normal cognition. In animal models, exposure to either chemotherapy or radiation adversely affects performance on tests of cognition specifically dependent on these brain systems, especially the hippocampus (Kim etal. , J. Radiat. Res. 49:517-526, 2008; Yang et al, Neurobiol. Learning and Mem. 93:487-494, 2010). Thus, drugs targeting these cortical and hippocampal systems could be neuroprotective in patients receiving cancer therapies and efficacious in treating symptoms of cognitive impairment that may last beyond the interventions used as cancer therapies.
[0187] Animal models serve as an important resource for developing and evaluating treatments for cancer therapy-related cognitive impairment. Features that characterize cancer therapy-related cognitive impairment in animal models typically extend to cancer therapy-related cognitive impairment in humans. Thus, efficacy in such animal models is expected to be predictive of efficacy in humans. Various animal models of cancer therapy-related cognitive impairment are known in the art. [0188] Examples of animal models of cancer therapy -related cognitive impairment include treating animals with anti -neoplastic agents such as cyclophosphamide (CYP) or with radiation, e.g., 60Co gamma-rays. (Kim et al., ./. Radiat. Res. 49:517-526, 2008; Yang et al, Neurobiol. Learning and Mem. 93:487-494, 2010). The cognitive function of animal models of cancer therapy-related cognitive impairment may then be tested with cognitive tests to assay the effectiveness of the methods and compositions of the invention in treating cancer therapy-related cognitive impairment. The efficacy of the methods and compositions of this invention in treating cancer therapy-related cognitive impairment, as well as human subjects with cancer therapy-related cognitive impairment, using a variety of cognitive tests known in the art, as discussed herein.
Parkinson’s disease (PD)
[0189] Parkinson’s disease (PD) is a neurological disorder characterized by a decrease of voluntary movements. The afflicted patient has reduction of motor activity and slower voluntary movements compared to the normal individual. The patient has characteristic "mask" face, a tendency to hurry while walking, bent over posture and generalized weakness of the muscles. There is a typical "lead-pipe" rigidity of passive movements. Another important feature of the disease is the tremor of the extremities occurring at rest and decreasing during movements.
[0190] Parkinson’s disease psychosis is experienced by about one third of PD patients and significantly affects the patient’s quality of life. Psychosis is characterized by hallucinations, delusions, and other sensory disturbances including illusions and “sense of presence” hallucinations. The underlying cause of psychosis in PD patients is not well understood. However, the occurrence of cognitive impairment in PD patients has been identified as a risk factor associated with the development of psychosis (Laura B. Zahodne and Hubert H. Fernandez, Drugs Aging. 2008, 25(8), 665-682).
[0191] Parkinson's disease, the etiology of which is unknown, belongs to a group of the most common movement disorders named parkinsonism, which affects approximately one person per one thousand. These other disorders grouped under the name of parkinsonism may result from viral infection, syphilis, arteriosclerosis and trauma and exposure to toxic chemicals and narcotics. Nonetheless, it is believed that the inappropriate loss of synaptic stability may lead to the disruption of neuronal circuits and to brain diseases. Whether as the result of genetics, drug use, the aging process, viral infections, or other various causes, dysfunction in neuronal communication is considered the underlying cause for many neurologic diseases, such as PD (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10, 207-214).
[0192] Regardless of the cause of the disease, the main pathologic feature is degeneration of dopaminergic cells in basal ganglia, especially in substantia nigra. Due to premature death of the dopamine containing neurons in substantia nigra, the largest structure of the basal ganglia, the striatum, will have reduced input from substantia nigra resulting in decreased dopamine release. The understanding of the underlying pathology led to the introduction of the first successful treatment which can alleviate Parkinson's disease. Virtually all approaches to the therapy of the disease are based on dopamine replacement. Drugs currently used in the treatment can be converted into dopamine after crossing the blood brain barrier, or they can boost the synthesis of dopamine and reduce its breakdown. Unfortunately, the main pathologic event, degeneration of the cells in substantia nigra, is not helped. The disease continues to progress and frequently after a certain length of time, dopamine replacement treatment will lose its effectiveness.
[0193] The invention provides methods and compositions for treating PD using a a5- containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of PD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with PD. In certain embodiments, the symptom to be treated is cognitive impairment. For example, methods and compositions of the disclosure can be used to improve the motor/cognitive impairments symptomatic of Parkinson’s disease. Moreover, methods and compositions of the disclosure may be useful for treating the memory impairment symptomatic of Parkinson’s disease. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with PD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In another embodiment of the invention, there is provide a method of treating Parkinson’s disease psychosis, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer or combination thereof.
[0194] There are a number of animal models for PD. Exemplary animal models for PD include the reserpine model, the methamphetamine model, the 6-hydroxydopamine (6- OHDA) model, the l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) model, the paraquat (PQ)-Maneb model, the rotenone model, the 3-nitrotyrosine model and genetic models using transgenic mice. Transgenic models include mice that over express a- synuclein, express human mutant forms of a -synuclein, or mice that express LRKK2 mutations. See review of these models by Ranjita B. et al. (Ranjita B. et al. BioEssays 2002, 24, 308-318). Additional information regarding these animal models is readily available from Jackson Laboratories (see also http://research.jax.org/grs/parkinsons.html), as well as in numerous publications disclosing the use of these validated models.
[0195] The efficacy of the methods and compositions of this invention in treating PD, or cognitive impairment associated with PD, may be assessed in any of the above animal models of PD, as well as human subjects with PD, using a variety of cognitive tests known in the art, as discussed herein.
Autism
[0196] Autism is a neurodevelopmental disorder characterized by dysfunction in three core behavioral dimensions: repetitive behaviors, social deficits, and cognitive deficits.
The repetitive behavior domain involves compulsive behaviors, unusual attachments to objects, rigid adherence to routines or rituals, and repetitive motor mannerisms such as stereotypies and self- stimulatory behaviors. The social deficit dimension involves deficits in reciprocal social interactions, lack of eye contact, diminished ability to carry on conversation, and impaired daily interaction skills. The cognitive deficits can include language abnormalities. Autism is a disabling neurological disorder that affects thousands of Americans and encompasses a number of subtypes, with various putative causes and few documented ameliorative treatments. The disorders of the autistic spectrum may be present at birth, or may have later onset, for example, at ages two or three. There are no clear cut biological markers for autism. Diagnosis of the disorder is made by considering the degree to which the child matches the behavioral syndrome, which is characterized by poor communicative abilities, peculiarities in social and cognitive capacities, and maladaptive behavioral patterns. The dysfunction in neuronal communication is considered one of the underlying causes for autism (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10, 207-214). Recent studies have shown that there is a GABAA a5 deficit in autism spectrum disorder (ASD) and support further investigations of the GABA system in this disorder (Mendez MA, et al. Neuropharmacology. 2013, 68:195-201).
[0197] The invention also provides methods and compositions for treating autism using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of autism. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with autism. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive deficit. For example, methods and compositions of the disclosure can be used to improve the motor/cognitive deficits symptomatic of autism. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with autism, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0198] The valproic acid (VP A) rat model of autism using in vitro electrophysiological techniques, established by Rodier et al. (Rodier, P. M. et al. Reprod. Toxicol. 1997, 11, 417-422) is one of the most exhaustively established insult-based animal models of autism and is based on the observation that pregnant women treated with VPA in the 1960s, during a circumscribed time window of embryogenesis, had a much higher risk of giving birth to an autistic child than the normal population. Offspring of VP A-exposed pregnant rats show several anatomical and behavioral symptoms typical of autism, such as diminished number of cerebellar Purkinje neurons, impaired social interaction, repetitive behaviors as well as other symptoms of autism, including enhanced fear memory processing. See, Rinaldi T. et al. Frontiers in Neural Circuits, 2008, 2, 1-7. Another mouse model, BTBR T+tf/J (BTBR) mice, an established model with robust behavioral phenotypes relevant to the three diagnostic behavioral symptoms of autism— unusual social interactions, impaired communication, and repetitive behaviors — was used to probe the efficacy of a selective negative allosteric modulator of the mGluR5 receptor, GRN-529. See, e.g., Silverman J. L. et al. Sci Transl. Med. 2012, 4, 131. The efficacy of the methods and compositions of this invention in treating autism, or cognitive deficits associated with autism, may be assessed in the VPA-treated rat model of autism or the BTBR T+tf/J (BTBR) mouse model, as well as human subjects with autism, using a variety of cognitive tests known in the art, as discussed herein.
Mental retardation
[0199] Mental retardation is a generalized disorder characterized by significantly impaired cognitive function and deficits in adaptive behaviors. Mental retardation is often defined as an Intelligence Quotient (IQ) score of less than 70. Inborn causes are among many underlying causes for mental retardation. The dysfunction in neuronal communication is also considered one of the underlying causes for mental retardation (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10, 207-214).
[0200] In some instances, mental retardation includes, but are not limited to, Down syndrome, velocariofacial syndrome, fetal alcohol syndrome, Fragile X syndrome, Klinefelter’s syndrome, neurofibromatosis, congenital hypothyroidism, Williams syndrome, phenylketonuria (PKU), Smith-Lemli-Opitz syndrome, Prader-Willi syndrome, Phelan-McDermid syndrome, Mowat-Wilson syndrome, ciliopathy, Lowe syndrome and siderium type X-linked mental retardation. Down syndrome is a disorder that includes a combination of birth defects, including some degree of mental retardation, characteristic facial features and, often, heart defects, increased infections, problems with vision and hearing, and other health problems. Fragile X syndrome is a prevalent form of inherited mental retardation, occurring with a frequency of 1 in 4,000 males and 1 in 8,000 females. The syndrome is also characterized by developmental delay, hyperactivity, attention deficit disorder, and autistic-like behavior. There is no effective treatment for fragile X syndrome.
[0201] The present invention contemplates the treatment of mild mental retardation, moderate mental retardation, severe mental retardation, profound mental retardation, and mental retardation severity unspecified. Such mental retardation may be, but is not required to be, associated with chromosomal changes, (for example Down Syndrome due to trisomy 21), heredity, pregnancy and perinatal problems, and other severe mental disorders. This invention provides methods and compositions for treating mental retardation using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of mental retardation. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with mental retardation. In certain embodiments, the symptom to be treated is cognitive deficit/impairment. For example, methods and compositions of the disclosure can be used to improve the motor/cognitive impairments symptomatic of mental retardation. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with mental retardation, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0202] Several animal models have been developed for mental retardation. For example, a knockout mouse model has been developed for Fragile X syndrome. Fragile X syndrome is a common form of mental retardation caused by the absence of the FMR1 protein, FMRP. Two homologs of FMRP have been identified, FXR1P and FXR2P. FXR2P shows high expression in brain and testis, like FMRP. Both Fxr2 and Fmrl knockout mice, and Fmrl/Fxr2 double knockout mice are believed to be useful models for mental retardation such as Fragile X syndrome. See, Bontekoe C. J. M. et al. Hum. Mol. Genet. 2002, 11 (5): 487-498. The efficacy of the methods and compositions of this invention in treating mental retardation, or cognitive deficit/impairment associated with mental retardation, may be assessed in the these mouse models and other animal models developed for mental retardation, as well as human subjects with mental retardation, using a variety of cognitive tests known in the art, as discussed herein.
Compulsive behavior (obsessive-compulsive disorder)
[0203] Obsessive compulsive disorder ("OCD") is a mental condition that is most commonly characterized by intrusive, repetitive unwanted thoughts (obsessions) resulting in compulsive behaviors and mental acts that an individual feels driven to perform (compulsion). Current epidemiological data indicates that OCD is the fourth most common mental disorder in the United States. Some studies suggest the prevalence of OCD is between one and three percent, although the prevalence of clinically recognized OCD is much lower, suggesting that many individuals with the disorder may not be diagnosed. Patients with OCD are often diagnosed by a psychologist, psychiatrist, or psychoanalyst according to the Diagnostic and Statistical Manual of Mental Disorders, 4th edition text revision (DSM-IV-TR) (2000) diagnostic criteria that include characteristics of obsessions and compulsions. Characteristics of obsession include: (1) recurrent and persistent thoughts, impulses, or images that are experienced as intrusive and that cause marked anxiety or distress; (2) the thoughts, impulses, or images are not simply excessive worries about real-life problems; and (3) the person attempts to ignore or suppress such thoughts, impulses, or images, or to neutralize them with some other thought or action.
The person recognizes that the obsessional thoughts, impulses, or images are a product of his or her own mind, and are not based in reality. Characteristics of compulsion include: (1) repetitive behaviors or mental acts that the person feels driven to perform in response to an obsession, or according to rules that must be applied rigidly; (2) the behaviors or mental acts are aimed at preventing or reducing distress or preventing some dreaded event or situation; however, these behaviors or mental acts are not actually connected to the issue, or they are excessive.
[0204] Individuals with OCD typically perform tasks (or compulsion) to seek relief from obsession-related anxiety. Repetitive behaviors such as handwashing, counting, checking, or cleaning are often performed with the hope of preventing obsessive thoughts or making them go away. Performing these "rituals," however, only provides temporary relief. People with OCD may also be diagnosed with a spectrum of other mental disorders, such as generalized anxiety disorder, anorexia nervosa, panic attack, or schizophrenia.
[0205] The dysfunction in neuronal communication is considered one of the underlying causes for obsession disorder (Myrrhe van Spronsen and Casper C. Hoogenraad, Curr. Neurol. Neurosci. Rep. 2010, 10, 207-214). Studies suggest that OCD may be related to abnormal levels of a neurotransmitter called serotonin. The first-line treatment of OCD consists of behavioral therapy, cognitive therapy, and medications. Medications for treatment include serotonin reuptake inhibitors (SRIs) such as paroxetine (Seroxat™, Paxil®, Xetanor™, ParoMerck™, Rexetin™), sertraline (Zoloft®, Stimuloton™), fluoxetine (Prozac®, Bioxetin™), escitalopram (Lexapro®), and fluvoxamine (Luvox®) as well as the tricyclic antidepressants, in particular clomipramine (Anafranil®). Benzodiazepines are also used in treatment. As much as 40 to 60% of the patients, however, fail to adequately respond to the SRI therapy and an even greater proportion of patients fail to experience complete remission of their symptoms.
[0206] The invention provides methods and compositions for treating OCD using a a5- containing GABAA receptor agonist (e.g, a a5-containing GABAA receptor positive allosteric modulator), such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of OCD. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with OCD. In certain embodiments, the symptom to be treated is cognitive impairment or cognitive deficit. For example, methods and compositions of the disclosure can be used to treat the cognitive deficits in OCD, and/or to improve cognitive function in patients with OCD. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with OCD, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0207] A quinpirole-sensitized rat model has been developed for OCD. The compulsive checking behavior of the quinpirole-sensitized rats is subject to interruption, which is an attribute characteristic of OCD compulsions. In addition, a schedule-induced polydipsia (SIP) rodent model of obsessive-compulsive disorder was used to evaluate the effects of the novel 5-HT2C receptor agonist WAY-163909. See, e.g, Rosenzweig-Lipson S. et al. Psychopharmacology (Berl) 2007, 192, 159-70. The efficacy of the methods and compositions of this invention in treating OCD, or cognitive impairment or cognitive deficits associated with OCD, may be assessed in the above animal models and other animal models developed for OCD, as well as human subjects with OCD, using a variety of cognitive tests known in the art, as discussed herein.
Substance addiction
[0208] Substance addiction (e.g, drug substance addiction, alcohol substance addiction) is a mental disorder. The substance addiction is not triggered instantaneously upon exposure to substance of abuse. Rather, it involves multiple, complex neural adaptations that develop with different time courses ranging from hours to days to months (Kauer J. A. Nat. Rev. Neurosci. 2007, 8, 844-858). The path to substance addiction generally begins with the voluntary use of one or more controlled substances, such as narcotics, barbiturates, methamphetamines, alcohol, nicotine, and any of a variety of other such controlled substances. Over time, with extended use of the controlled substance(s), the voluntary ability to abstain from the controlled substance(s) is compromised due to the effects of prolonged use on brain function, and thus on behavior. As such, substance addiction generally is characterized by compulsive substance craving, seeking and use that persist even in the face of negative consequences. The cravings may represent changes in the underlying neurobiology of the patient which likely must be addressed in a meaningful way if recovery is to be obtained. Substance addiction is also characterized in many cases by withdrawal symptoms, which for some substances are life threatening ( e.g ., alcohol, barbiturates) and in others can result in substantial morbidity (which may include nausea, vomiting, fever, dizziness, and profuse sweating), distress, and decreased ability to obtain recovery. For example, alcoholism, also known as alcohol dependence, is one such substance addiction. Alcoholism is primarily characterized by four symptoms, which include cravings, loss of control, physical dependence and tolerance. These symptoms also may characterize substance addictions to other controlled substances. The craving for alcohol, as well as other controlled substances, often is as strong as the need for food or water. Thus, an alcoholic may continue to drink despite serious family, health and/or legal ramifications.
[0209] Recent work exploring the effects of abusing alcohol, central stimulants, and opiates on the central nervous system (CNS) have demonstrated a variety of adverse effects related to mental health, including substance-induced impairments in cognition.
See, Nyberg F. Cognitive Impairments in Drug Addicts, Chapter 9. In several laboratories and clinics substantial damages of brain function are seen to result from these drugs. Among the harmful effects of the abusing drugs on brain are those contributing to accelerated obsolescence. An observation that has received special attention during recent years is that chronic drug users display pronounced impairment in brain areas associated with executive and memory function. A remarked neuroadaptation caused by addictive drugs, such as alcohol, central stimulants and opiates involves diminished neurogenesis in the subgranular zone (SGZ) of the hippocampus. Indeed, it has been proposed that decreased adult neurogenesis in the SGZ could modify the hippocampal function in such a way that it contributes to relapse and a maintained addictive behavior. It also raises the possibility that decreased neurogenesis may contribute to cognitive deficits elicited by these abusing drugs.
[0210] The invention provides methods and compositions for treating substance addiction using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of substance addiction. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with substance addiction. In certain embodiments, the symptom to be treated is cognitive impairment. For example, methods and compositions of the disclosure can be used to treat the cognitive impairment and/or to improve cognitive function in patients with substance addiction. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with substance addiction, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
[0211] Several animal models have been developed to study substance addiction. For example, a genetically selected Marchigian Sardinian alcohol-preferring (msP) rat models was developed to study the neurobiology of alcoholism. See, Ciccocioppo R. et al. Substance addiction Biology 2006, 11, 339-355. The efficacy of the methods and compositions of this invention in treating substance addiction, or cognitive impairment associated with substance addiction, may also be assessed in animal models of substance addiction, as well as human subjects with substance addiction, using a variety of cognitive tests known in the art, as discussed herein.
Brain Cancers
[0212] Brain cancer is the growth of abnormal cells in the tissues of the brain usually related to the growth of malignant brain tumors. Brain tumors grow and press on the nearby areas of the brain which can stop that part of the brain from working the way it should. Brain cancer rarely spreads into other tissues outside of the brain. The grade of tumor, based on how abnormal the cancer cells look under a microscope, may be used to tell the difference between slow- and fast-growing tumors. Brain tumors are classified according to the kind of cell from which the tumor seems to originate. Diffuse, fibrillary astrocytomas are the most common type of primary brain tumor in adults.
These tumors are divided histopathologically into three grades of malignancy: World Health Organization (WHO) grade II astrocytoma, WHO grade III anaplastic astrocytoma and WHO grade IV glioblastoma multiforme (GBM). WHO grade II astocytomas are the most indolent of the diffuse astrocytoma spectrum. Astrocytomas display a remarkable tendency to infiltrate the surrounding brain, confounding therapeutic attempts at local control. These invasive abilities are often apparent in low-grade as well as high- grade tumors.
[0213] Glioblastoma multiforme is the most malignant stage of astrocytoma, with survival times of less than 2 years for most patients. Histologically, these tumors are characterized by dense cellularity, high proliferation indices, endothelial proliferation and focal necrosis. The highly proliferative nature of these lesions likely results from multiple mitogenic effects. One of the hallmarks of GBM is endothelial proliferation. A host of angiogenic growth factors and their receptors are found in GBMs.
[0214] There are biologic subsets of astrocytomas, which may reflect the clinical heterogeneity observed in these tumors. These subsets include brain stem gliomas, which are a form of pediatric diffuse, fibrillary astrocytoma that often follow a malignant course. Brain stem GBMs share genetic features with those adult GBMs that affect younger patients. Pleomorphic xanthoastrocytoma (PXA) is a superficial, low-grade astrocytic tumor that predominantly affects young adults. While these tumors have a bizarre histological appearance, they are typically slow-growing tumors that may be amenable to surgical cure. Some PXAs, however, may recur as GBM. Pilocytic astrocytoma is the most common astrocytic tumor of childhood and differs clinically and histopathologically from the diffuse, fibrillary astrocytoma that affects adults. Pilocytic astrocytomas do not have the same genomic alterations as diffuse, fibrillary astrocytomas. Subependymal giant cell astrocytomas (SEGA) are periventricular, low-grade astrocytic tumors that are usually associated with tuberous sclerosis (TS), and are histologically identical to the so-called “candle-gutterings” that line the ventricles of TS patients. Similar to the other tumorous lesions in TS, these are slowly-growing and may be more akin to hamartomas than true neoplasms. Desmoplastic cerebral astrocytoma of infancy (DCAI) and desmoplastic infantile ganglioglioma (DIGG) are large, superficial, usually cystic, benign astrocytomas that affect children in the first year or two of life.
[0215] Oligodendrogliomas and oligoastrocytomas (mixed gliomas) are diffuse, usually cerebral tumors that are clinically and biologically most closely related to the diffuse, fibrillary astrocytomas. The tumors, however, are far less common than astrocytomas and have generally better prognoses than the diffuse astrocytomas. Oligodendrogliomas and oligoastrocytomas may progress, either to WHO grade III anaplastic oligodendroglioma or anaplastic oligoastrocytoma, or to WHO grade IV GBM. Thus, the genetic changes that lead to oligodendroglial tumors constitute yet another pathway to GBM.
[0216] Ependymomas are a clinically diverse group of gliomas that vary from aggressive intraventricular tumors of children to benign spinal cord tumors in adults. Transitions of ependymoma to GBM are rare. Choroid plexus tumors are also a varied group of tumors that preferentially occur in the ventricular system, ranging from aggressive supratentorial intraventricular tumors of children to benign cerebellopontine angle tumors of adults. Choroid plexus tumors have been reported occasionally in patients with Li-Fraumeni syndrome and von Hippel-Lindau (VHL) disease.
[0217] Medulloblastomas are highly malignant, primitive tumors that arise in the posterior fossa, primarily in children. Medulloblastoma is the most common childhood malignant brain tumor. The most lethal medulloblastoma subtype exhibits a high expression of the GABAA receptor a5 subunit gene and MYC amplification. See, e.g., J Biomed Nanotechnol. 2016 Jun; 12(6): 1297-302.
[0218] Meningiomas are common intracranial tumors that arise in the meninges and compress the underlying brain. Meningiomas are usually benign, but some “atypical” meningiomas may recur locally, and some meningiomas are frankly malignant and may invade the brain or metastasize. Atypical and malignant meningiomas are not as common as benign meningiomas. Schwannomas are benign tumors that arise on peripheral nerves. Schwannomas may arise on cranial nerves, particularly the vestibular portion of the eighth cranial nerve (vestibular schwannomas, acoustic neuromas) where they present as cerebellopontine angle masses. Hemangioblastomas are tumors of uncertain origin that are composed of endothelial cells, pericytes and so-called stromal cells. These benign tumors most frequently occur in the cerebellum and spinal cord of young adults. Multiple hemangioblastomas are characteristic of von Hippel-Lindau disease (VHL). Hemangiopericytomas (HPCs) are dural tumors which may display locally aggressive behavior and may metastasize. The histogenesis of dural-based hemangiopericytoma (HPC) has long been debated, with some authors classifying it as a distinct entity and others classifying it as a subtype of meningioma.
[0219] The invention provides methods and compositions for treating brain cancers (for example, brain tumors as described herein) using a a5-containing GABAA receptor positive allosteric modulator, such as one selected from the compounds or pharmaceutically acceptable salts, hydrates, solvates, polymorphs, isomers, or combinations thereof as described herein. In certain embodiments, treatment comprises preventing or slowing the progression of brain cancers. In certain embodiments, treatment comprises alleviation, amelioration, or slowing the progression of one or more symptoms associated with brain cancers. In certain embodiments, the symptom to be treated is cognitive impairment. For example, methods and compositions of the disclosure can be used to treat the cognitive impairment and/or to improve cognitive function in patients with brain cancers. In some embodiments of the invention, there is provided a method of preserving or improving cognitive function in a subject with brain cancers, the method comprising the step of administering to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. In some embodiments, the brain tumor is medulloblastoma.
Research Domain Criteria (RDoC)
[0220] The invention further provides methods and compositions for treating impairment in neurological disorders and neuropsychiatric conditions using a a5- containing GABAA R positive allosteric modulator or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof as described herein. In certain embodiments, treatment comprises alleviation, amelioration or slowing the progression, of one or more symptoms associated with such impairment. In another aspect of the invention, there is provided methods and compositions for preserving or improving cognitive function in a subject in need thereof using a compound of the invention or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof. [0221] Research Domain Criteria (RDoC) are expected to augment clinical criteria, such as DSM and ICD, for diagnosis of disease and disorders affecting the nervous system (see, e.g., Am. J. Psychiatry 167:7 (2010)). The RDoC is intended to provide classification based on discoveries in genomics and neuroscience as well as clinical observation. The high expression of a5 -containing GABAA receptors in specific neural circuits in the nervous system could be therapeutic targets for neural circuit dysfunction identified under RDoC.
Assays for GABAA a5 subunit binding and receptor positive allosteric modulator activity
[0222] The affinity of test compounds for a GABAA receptor comprising the GABAA a5 subunit may be determined using receptor binding assays that are known in the art. See, e.g., U.S. Patent 7,642,267 and U.S. Patent 6,743,789, which are incorporated herein by reference.
[0223] The activity of the test compounds as a a5 -containing GABAA R positive allosteric modulator may be tested by electrophysiological methods known in the art. See, e.g., U.S. Patent 7,642,267 and Guidotti et al, Psychopharmacology 180: 191-205, 2005. Positive allosteric modulator activity may be tested, for examples, by assaying GABA- induced chloride ion conductance of GABAA receptors comprising the GABAA a5 subunit. Cells expressing such receptors may be exposed to an effective amount of a compound of the invention. Such cells may be contacted in vivo with compounds of the invention through contact with a body fluid containing the compound, for example through contact with cerebrospinal fluid. In vitro tests may be done by contacting cells with a compound of the invention in the presence of GABA. Increased GABA-induced chloride conductance in cells expressing GABAA receptors comprising the GABAA a5 subunit in the presence of the test compound would indicate positive allosteric modulator activity of said compound. Such changes in conductance may be detected by, e.g, using a voltage-clamp assay performed on Xenopus oocytes injected with GABAA receptor subunit mRNA (including GABAA a5 subunit RNA), HEK 293 cells transfected with plasmids encoding GABAA receptor subunits, or in vivo, ex vivo, or cultured neurons.
[0224] It will be understood by one of ordinary skill in the art that the methods described herein may be adapted and modified as is appropriate for the application being addressed and that the methods described herein may be employed in other suitable applications, and that such other additions and modifications will not depart from the scope hereof.
[0225] The compounds of this disclosure can by synthesized using analogous methods to those described in WO 2018/130868. However, one skilled in the art will readily appreciate that the specific methods and results discussed are merely illustrative of the invention as described more fully in the embodiments which follow thereafter.
[0226] Compounds 731 and 736 were prepared by subjecting Compound 328 of WO 2018/130868 to Sonogashira reaction conditions using appropriate starting materials. For exemplary reaction conditions, see Scheme 29 and Compound 285 of WO 2018/130868.
[0227] Compounds 733-734 were prepared by subjecting Compound 356 (Scheme 41) of WO 2018/130868 to Sonogashira reaction conditions using the appropriate starting materials. For exemplary reaction conditions, see Scheme 29 and Compound 285 of WO 2018/130868.
[0228] Compounds 732 and 735 were prepared by subjecting Compound 403 of WO 2018/130868 to Sonogashira reaction conditions using the appropriate starting materials. For exemplary reaction conditions, see Scheme 29 and Compound 285 of WO 2018/130868.
[0229] Compound 737 was prepared by subjecting Compound 256 (Scheme 29) of WO 2018/130868 to Sonogashira reaction conditions using the appropriate starting materials. For exemplary reaction conditions, see Scheme 29 and Compound 285 of WO 2018/130868.
[0230] Synthesis of Compound 633
633
Scheme 11
[0231] To a stirred solution of compound 17 (see Scheme 11 of WO 2018/130868, 17: R1=OMe; 17’: R^Cl) (2.96 g, 11.2 mmol), imidazole (1.91 g, 28.1 mmol), DMAP (274 mg, 2.24 mmol), and Et3N (4.7 mL, 33.7 mmol) in anhydrous CH2CI2 (25 mL) and anhydrous DMF (25 mL) was added TBSC1 (3.37 g, 22.4 mmol). The reaction mixture was stirred at rt under N2 for overnight. After this time, the resulting reaction mixture was diluted with EtOAc (300 mL), washed with 10% LiCl aqueous solution (3 x 50 mL) and brine (50 mL), dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 0% to 5% EtOAc/CLhCh to provide compound 633-1 as a white solid (3.67 g, 87%) : MS [M+Na] = 401. [0232] To a stirred solution of compound 633-1 (3.67 g, 9.69 mmol) in anhydrous DMF (50 mL) was added NaH (60% in mineral oil, 426 mg, 10.7 mmol) at -20 °C under N2.
[0233] The resulting mixture was stirred at -20 °C for 10 min. After this time, PMBC1 (2.0 mL, 14.8 mmol) was added. The reaction mixture was warmed to rt overnight. The resulting mixture was quenched with 10% LiCl aqueous solution (100 mL) and extracted with EtOAc (3 cx 100 mL). The combined extracts were washed with 10% LiCl aqueous solution (3 x 30 mL) and brine (50 mL), dried over anhydrous NaiSCL, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 10% to 30% EtO Ac/hexanes to afford compound 633-2 as a light yellow solid (4.59 g, 95%): MS [M+l] = 499.
[0234] To a stirred solution of compound 633-2 (4.59 g, 9.20 mmol) in anhydrous THF (100 mL) was added TBAF (1 M solution in THF, 18.4 mL, 18.4 mmol) at 0 °C under N2. The reaction mixture was stirred at 0 °C for 2 h. After this time, the reaction was quenched with saturated NH4CI aqueous solution (100 mL) and extracted with CH2CI2 (3 x 100 mL). The combined extracts were dried over anhydrous INfeSCL, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 50% to 100% EtO Ac/hexanes to afford compound 633-3 as a white foam (2.92 g, 83%): MS [M+l] = 385.
[0235] To a stirred solution of compound 633-3 (2.92 g, 7.59 mmol) in anhydrous CH2CI2 (150 mL) was added PPt (3.98 g, 15.2 mmol) followed by CB (3.02 g, 9.11 mmol) at rt under N2. The reaction mixture was stirred at rt for 4 h. After this time, the reaction mixture was concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 30% to 60%
EtO Ac/hexanes to afford compound 633-4 as a white foam (2.59 g, 76%) : MS [M+l] = 447.
[0236] To a stirred solution of compound 633-4 (576 mg, 1.29 mmol) in anhydrous THF (10 mL) and anhydrous MeOH (45 mL) was added NaH (60% in mineral oil, 515 mg,
12.9 mmol) portion-wise over 40 min at 0 °C under N2. The reaction mixture was stirred at 0 °C for 1.5 h. After this time, the reaction was quenched with saturated NH4CI aqueous solution (100 mL) and neutralized by 6 N HC1 aqueous solution to pH ~7. The resulting mixture was extracted with CH2CI2 (3 x 100 mL). The combined extracts were dried over anhydrous INfeSCri, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 50% to 100% EtOAc/hexanes to afford compound 633-5 as a white foam (436 mg, 85%): MS [M+l] = 399.
[0237] A solution of compound 633-5 (1.66 g, 4.16 mmol) in TFA (15 mL) was heated to reflux (85 °C oil bath) for 2 days. After this time, the reaction mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in CH2CI2 (100 mL), washed with saturated NaHCCh (3 x 50 mL), dried over anhydrous Na2SC>4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 50% to 100% EtOAc/hexanes to afford compound 633-6 as an off-white solid (810 mg, 70%.): MS [M+l] = 279.
[0238] To a stirred solution of 1,2,4-triazole (506 mg, 7.33 mmol) in anhydrous CFLCN (30 mL) was added DIPEA (1.3 mL, 7.46 mmol) followed by POCb (0.2 mL, 2.15 mmol) at 0 °C under N2. The reaction mixture was stirred at 0 °C for 2 h. After this time, a solution of compound 633-6 (1.02 g, 3.66 mmol) in anhydrous CH3CN (50 mL) was added. The reaction mixture was warmed to rt, then heated to reflux (100 °C oil bath) for overnight. The reaction mixture was cooled to rt and quenched with ice cold water (50 mL). The resulting mixture was extracted with CH2CI2 (3 x 100 mL). The combined extracts were dried over anhydrous Na2SC>4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 50% to 100% EtOAc/CFhCh to afford compound 633-7 as a yellow solid (950 mg, 79%) : MS [M+l] = 330.
[0239] To a stirred solution of KOt-Bu (485 mg, 4.32 mmol) in anhydrous DMF (10 mL) was added CNCFhCCLEt (0.47 mL, 4.30 mmol) at -50 °C under N2. The reaction mixture was stirred at -50 °C for 1 h. After this time, a solution of compound 633-7 (950 mg, 2.88 mmol) in anhydrous DMF (15 mL) was added. The reaction mixture was slowly warmed to rt overnight. The reaction mixture was quenched with saturated NaHCCh aqueous solution (50 mL). The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined extracts were washed with 10% LiCl aqueous solution (3 x 30 mL) and brine (30 mL), dried over anhydrous Na2SC>4, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 0% to 8% MeOH/EtOAc to afford compound 633-8 as a yellow solid (815 mg, 76%) : MS [M+l] = 374.
[0240] To a stirred solution of compound 633-8 (150 mg, 0.401 mmol) in THF (15 mL) and water (7 mL) was added LiOTbThO (84 mg, 2.00 mmol) at rt. The reaction mixture was stirred at rt for 3 h. After this time, the reaction mixture was concentrated under reduced pressure. The resulting mixture was acidified with 2 N HC1 aqueous solution to pH~2. The solid was collected by filtration. The filter cake was washed with water (10 mL), dried under high vacuum to afford compound 633-9 as a white solid (125 mg, 90%): MS [M+l] = 346.
[0241] A suspension of compound 633-9 (122 mg, 0.353 mmol), h (269 mg, 1.06 mmol), and K3PO4 (75 mg, 0.353 mmol) in anhydrous DMF (9 mL) was sealed and placed in a microwave reactor at 170 °C for 30 min. After this time, the reaction mixture was cooled to rt and diluted with 10% LiCl aqueous solution (50 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined extracts were washed with 10% LiCl aqueous solution (3 x 30 mL) and brine (30 mL), dried over anhydrous NaiSCL, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 1% to 4% MeOH/CFbCb to afford compound 633-10 as a white solid (35 mg, 23%): MS [M+l] = 428.
[0242] A suspension of compound 633-10 (32 mg, 0.0748 mmol), compound 633-11 (37 pL, 0.303 mmol), and Cul (4 mg, 0.0210 mmol) in anhydrous DMF (4 mL) was bubbled with argon for 5 min. After this time, Et3N (52 pL, 0.373 mmol) was added followed by Pd(dppf)Cl2*CH2Cl2 (12 mg, 0.0147 mmol). The resulting mixture was heated at 40 °C under argon for 2 h. The reaction mixture was then cooled to rt, diluted with water (50 mL), and extracted with EtOAc (3 x 30 mL). The combined extracts were washed with 10% LiCl aqueous solution (3 x 20 mL) and brine (20 mL), dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure. The resulting residue was purified by flash column chromatography on silica gel eluting with 0% to 4% MeOFl/CFhCh to afford compound 633 as an off-white solid (13 mg, 41%): ¾ NMR (300 MHz, DMSO- is) d 8.45 (d, 7= 1.8 Hz, 1H), 8.27 (s, 1H), 8.13 (d, J= 2.3 Hz, 1H), 8.01 (d, J= 8.7, 2.3 Hz, 1H), 7.92 (dd, J= 8.6, 2.3 Hz, 1H), 7.86 (dd, J= 8.7, 2.3 Hz, 1H), 6.92 (d, J= 8.6 Hz, 1H), 4.64 (s, 2H), 4.43 (br s, 2H), 3.91 (s, 3H), 3.26 (s, 3H); ESI MS, m/z = 433 [M+H]+.
[0243] Compounds 738-740 were prepared by subjecting Compound 633-10 of Scheme 11 to Sonogashira reaction conditions, using the appropriate starting materials, under the conditions described for Compound 633 in Scheme 11.
[0244] Compounds 731-740 were characterized by MS and ¾ NMR. The MS characterization is summarized below in Table 5.
Table 5. MS characterization of Compounds 731-740:
Example 105: Assessing a5-containing GABAA Receptor (GABAAR) positive allosteric modulator activity
[0245] Step 1: Establish clones of GABAAR subunits (a5, [>3, y2, al, a2 and a3) and prepare the corresponding cRNAs Human clones of GABAA-R a5, b3, g2, al, a2 and a3 subunits are obtained from commercial resources ( e.g. , OriGene, http://www.origene.com and Genescript, http://www.genescript.com). These clones are engineered into pRC, pCDM, pcDNA, and pBluescript KSM vector (for oocyte expression) or other equivalent expression vectors. Conventional transfection agents (e.g, FuGene, Lipofectamine 2000, or others) are used to transiently transfect host cells.
[0246] Step 2 - Functional GABAAR Assay of </.5b3n2, a1b3g2, a2b3g2, and a3b3g2, subtypes inXenopus oocyte expression system. cRNAs encoding a5, b3, g2, al, a2 and a3 subunits are transcribed in vitro using T3 mMESSAGE mMACHINE Kit (Ambion) and injected (in a ratio of a:b:g = 2:2: 1 or other optimized conditions) into oocytes freshly prepared from Xenopus laevis. After two days of culturing, GABA-gated Cl- currents from oocytes are performed using TEVC setups (Warner Instruments, Inc., Foster City, CA). GABA, benzodiazepine, and diazepam are used as reference compounds to validate the system.
[0247] Step 3 - Evaluate test compounds for positive allosteric modulator activity on the a5b3g2 subtype and test off-target activity on the al to a3 coupled b3g2 subtypes when the EC50=5pM selectivity cut-off is reached. The GABA-gated Cl- current from oocytes are measured in the TEVC setup in the presence of the test compounds. The positive allosteric modulator activity of each the test compounds is tested in a 5-point dose- response assay. The test compounds include some reference compounds (literature EC50 values for the a5b3g2 subtype are in the range of 3-10 mM). EC50s in the a5b3g2 subtype are obtained for each compound. If the EC50 in a5b3g2 is < 5mM, then the EC50 of the other three subtypes (a1b2g2, a2b3g2, and a3b3g2) is further determined individually in order to test for selectivity of the compounds in the a5b3g2 subtype over other subtypes.
[0248] Step 4 Evaluate further test compounds on the a5b3g2 subtype and test off- target activities when the EC50=0.5pM selectivity cut-off is reached. The second batch of test compounds are tested using the same strategy, but with a lower EC50 cutoff (0.5 mM). Again, the EC50s of the a5b3g2 subtype for each of the compounds is determined. The al to a3 coupled b3g2 subtypes are tested only if the EC50 for the a5-containing receptor is < 0.5 mM.
Example 106: Evaluating Compounds for Binding and Positive Allosteric Modulator Activity on the GABAA a5 Receptors
(A) Binding activity of test compounds on GABAAR [0249] Tissue culture and Membrane Preparation: The binding was performed on Ltk cells stably expressing GABAA receptors: a1b1g2, a2b3g2, a3b3g2 and a5b3g2 (provided by Merck Co., NJ, USA). Cells were seeded in 100 mm culture plates in DMEM/F12 medium containing 10% serum and antibiotics in 5% C02 and allowed to grow for 1-2 days. GABAAR expression was then induced by dexamethasone as follows: 0.5 mM for 1 day for a5 containing and 2 mM for 3 days for al, a2 and a3 containing GABAARS. After induction, cells were collected by scraping into Dulbecco’s Phosphate buffered saline (DPBS, pH 7.4, Invitrogen, Carlsbad, CA, USA) and centrifuged at 150 x g for 10 min. The pellet was washed twice by re-suspension and centrifugation. The cell pellets from at least 5 different preps were combined, suspended in the binding assay buffer (50 mM KH2P04; 1 mM EDTA; 0.2 M KC1, pH 7.4) and membranes prepared by sonication (3-5 times, 30 sec) using Branson Sonifier 150 (G.Heinmann, Germany). Protein content was determined using BCA assay (Bio-Rad Labs, Reinach, Switzerland) with Bovine Serum Albumin (Sigma Aldrich, St. Louis, MO, USA) as the standard. Aliquots were prepared and stored at -20°C for further use in binding assays.
[0250] Ligand Binding: Saturation binding curves were obtained by incubating membranes with increasing concentrations (0.01 - 8 nM) of [¾]Rol5-1788 (Flumazepil, 75-85 Ci/mmol, PerkinElmer, MA, USA), with nonspecific binding measured in the presence of 10 mM diazepam. Inhibition of [3H]Rol5-1788 binding of the test compounds was performed at concentrations of the radioligand at or lower than the Kd values for al, a2, a3 and a5 containing GABAARS determined from the saturation curves.
[0251] All binding assays were performed for 1 h at 4°C in assay buffer. The total assay volume was 0.5 ml containing 0.2 mg/ml protein for a5 and 0.4 mg/ml for al, a2, and a3 containing GABAAR membranes. Incubations were terminated by filtration through GF/B filters using a 24-Cell Harvestor (Brandel, Gaithersburg, MD, USA) followed by 3 washes with ice-cold assay buffer. Filters were transferred to scintillation vials, 5 ml scintillation liquid added, vortex-mixed and kept in dark. Next day, radioactivity was obtained using a scintillation counter (Beckman Coulter, Brea, CA, USA). All assays were performed in triplicate.
[0252] Data Analyses: Saturation and inhibition curves were obtained using GraphPad Prism software (GraphPad Software, Inc., CA, USA). The equilibrium dissociation constants (Ki values) of the unlabeled ligand were determined using Cheng-Prusoff equation Ki= IC50/ (1+S/Kd), where IC50 is the concentration of unlabeled ligand that inhibits 50% of [3H] ligand binding, S is the concentration of radioligand and Kd is the equilibrium dissociation constant of the radioactive ligand. A log range of the compounds (1 nM - 10 mM) was used to determine the Ki values which are presented as Mean ± SD from triplicate assays.
(o) positive allosteric modulator activity of test compounds on abpzyz subtype (JABAAK
[0253] Compounds of the present invention were initially screened at 100 nM for their ability to potentiate an EC 20 concentration of GABA in oocytes containing GABAA receptors (a5b2g2), using a protocol essentially similar to the one presented above.
[0254] On day 1, lng/32nL of GABAA a5b2g2 cDNA was injected into one oocyte.
Test starts on day 2. The cDNA injected to the oocytes was a mix of alpha, beta and gamma, their ratio is 1:1:10 (by weight) and the total weight of the mixed 3 subunits to be injected in one oocyte was lng in 32 nl volume. The injected oocytes can also be tested on day 3. In such case, the cDNA amount injected to the oocytes should be reduced by 20%.
[0255] Compounds of the present invention were tested using the following procedures. G02561 GABA dose-response
1). 8 oocytes were placed in 8 chambers of OpusXpress and superfused with Modified Barth’s Saline (MBS) at 3mL/min. Glass electrodes back-filled with 3M KC1 (0.5-3 megaohms) were used. Membrane potential of oocytes was voltage-clamped at -60mV.
2). Average EC20 GABA obtained from previous tests were applied for five-six times to stabilize oocytes. Oocytes were washed with MBS for 5-10 min between each GABA applications.
3). Run GABA dose-response to obtain EC 20 GABA value.
G02571 Control test (Diazepam or methyl 3,5-diphenylpyridazine-4-carboxylate)
1). New oocytes were used to run new test.
2). EC20 GABA were applied for five-six times to stabilize oocytes. Oocytes were washed with MBS for 5-10 min between each GABA applications.
3). EC20 GABA was applied to obtain current (IGABA). Oocytes were washed with MBS for 5-10 min.
4). ImM diazepam or methyl 3,5-diphenylpyridazine-4-carboxylate was pre-applied for 40 sec, followed by co-application of ImM diazepam or methyl 3,5-diphenylpyridazine-4- carboxylate and EC20 GABA to obtain Itest- best was divided by IGABA to obtain potentiation (%).
1). Repeat the above steps 1), 2) and 3) in the control test.
2). The first concentration of a test compound was pre-applied for 40 sec followed by co application of the test compound of the same concentration and EC20 GABA to obtain Itest. Divide best by IGABA to obtain potentiation (%).
3). Discard all tested oocytes, new oocytes were used and the above steps 1) and 2) were repeated to test second concentration of the same compound. Each oocyte was used for only one concentration test for a single test compound. The steps were repeated for other test compounds.
[0259] In some embodiments, the compounds of this application have a binding affinity (as represented by Ki) at a5-containing GABAARS of less than 200 nM, less than 180 nM, less than 150 nM, or less than 100 nM. In some embodiments, the compounds of this application have a binding affinity (as represented by Ki) at a5-containing GABAARS of less than 50 nM. In some embodiments, the compounds of this application have a binding affinity (as represented by Ki) at a5-containing GABAARS of less than 10 nM.
[0260] In some embodiments, the compounds of this application are selective for a5- containing GABAARS over al-containing GABAARS. In some embodiments, the compounds of this application are more than 50-fold, more than 100-fold, more than 500- fold or more than 1000-fold selective for a5-containing GABAARS over al-containing GABAARS.
[0261] In some embodiments, the compounds of this application have an EC50 at the a5- containing GABAARS of less than 500 nM, less than 100 nM or less than 50 nM. In some embodiments, the compounds of this application have an EC50 at the a5-containing GABAARS of less than 25 nM.
[0262] In some embodiments, the compounds of this application potentiate a5- containing GABAARS for more than 10%, more than 25%, more than 50%, or more than 75% at 100 nM. In some embodiments, the compounds of this application potentiate a5- containing GABAARS for more than 10%, more than 25%, more than 50%, or more than 75% at 1000 nM.
[0263] Screening results of the binding and PAM functional activity tests are summarized in Tables 1 and 2 below. [0264] The following Table 1 illustrates the ranges of GABA a.5 binding Ki’s associated with compounds of this disclosure: Table 1
[0265] The following Table 2 illustrates the ranges of GABA a.5 functional potentiation associated with compounds of this disclosure: Table 2
[0266] Selected compounds of this invention demonstrate > 10-fold binding selectivity for GABA a.5 versus GABA al, GABA a2, or GABA a.3. Some compounds of this application demonstrate over 20-fold, 50-fold, or 100-fold binding selectivity for GABA a.5 versus GABA a 1, GABA a2, or GABA a.3.
[0267] The following Table 6 illustrates the ranges of the binding selectivity of the compounds of the present disclosure for GABA a5 versus GABA al, GABA a2, or GABA a3:
Table 6
Example 107: Effect of Methyl 3,5-diphenylpyridazine-4-carboxylate in Aged- Impaired (Al) Rats
[0268] Methyl 3,5-diphenylpyridazine-4-carboxylate, corresponding to compound number 6 in van Niel et al. J. Med. Chem. 48:6004-6011 (2005), is a selective a5- containing GABAA R agonist. It has an a.5 in vitro efficacy of +27 (EC20). The effect of methyl 3,5-diphenylpyridazine-4-carboxylate in aged-impaired rats was studied using a RAM task. Moreover, receptor occupancy by methyl 3,5-diphenylpyridazine-4- carboxylate in a5-containing GABAA receptor was also studied.
(A) Effect of Methyl 3,5-divhenylvyridazine-4-carboxylate in Aged-Imyaired Rats U sins a Radial Arm Maze (RAM) Behavioral Task
[0269] The effects of methyl 3,5-diphenylpyridazine-4-carboxylate on the in vivo spatial memory retention of aged-impaired (Al) rats were assessed in a Radial Arm Maze (RAM) behavioral task using vehicle control and four different dosage levels of methyl 3,5- diphenylpyridazine-4-carboxylate (0.1 mg/kg, 0.3 mg/kg, 1 mg/kg and 3 mg/kg, ip). RAM behavioral tasks were performed on eight AI rats. All five treatment conditions (vehicle and four dosage levels) were tested on all eight rats.
[0270] The RAM apparatus used consisted of eight equidistantly-spaced arms. An elevated maze arm (7 cm width x 75 cm length) projected from each facet of an octagonal center platform (30 cm diameter, 51.5 cm height). Clear side walls on the arms were 10 cm high and were angled at 65° to form a trough. A food well (4 cm diameter, 2 cm deep) was located at the distal end of each arm. Froot Loops™ (Kellogg Company) were used as rewards. Blocks constructed of Plexiglas™ (30 cm height x 12 cm width) could be positioned to prevent entry to any arm. Numerous extra maze cues surrounding the apparatus were also provided.
[0271] The AI rats were initially subjected to a pre-training test (Chappell et al. Neuropharmacology 37: 481-487, 1998). The pre-training test consisted of a habituation phase (4 days), a training phase on the standard win-shift task (18 days) and another training phase (14 days) in which a brief delay was imposed between presentation of a subset of arms designated by the experimenter ( e.g. , 5 arms available and 3 arms blocked) and completion of the eight-arm win-shift task (/. ., with all eight arms available).
[0272] In the habituation phase, rats were familiarized to the maze for an 8-minute session on four consecutive days. In each of these sessions, food rewards were scattered on the RAM, initially on the center platform and arms and then progressively confined to the arms. After this habituation phase, a standard training protocol was used, in which a food pellet was located at the end of each arm. Rats received one trial each day for 18 days. Each daily trial terminated when all eight food pellets had been obtained or when either 16 choices were made or 15 minutes had elapsed. After completion of this training phase, a second training phase was carried out in which the memory demand was increased by imposing a brief delay during the trial. At the beginning of each trial, three arms of the eight-arm maze were blocked. Rats were allowed to obtain food on the five arms to which access was permitted during this initial “information phase” of the trial.
Rats were then removed from the maze for 60 seconds, during which time the barriers on the maze were removed, thus allowing access to all eight arms. Rats were then placed back onto the center platform and allowed to obtain the remaining food rewards during this “retention test” phase of the trial. The identity and configuration of the blocked arms varied across trials. [0273] The number of “errors” the AI rats made during the retention test phase was tracked. An error occurred in the trial if the rats entered an arm from which food had already been retrieved in the pre-delay component of the trial, or if the rat re-visited an arm in the post-delay session that it had already visited.
[0274] After completion of the pre-training test, rats were subjected to trials with more extended delay intervals, i.e., a two-hour delay, between the information phase (presentation with some blocked arms) and the retention test (presentation of all arms). During the delay interval, rats remained off to the side of the maze in the testing room, on carts in their individual home cages. AI rats were pretreated 30 - 40 minutes before daily trials with a one-time shot of the following five conditions: 1) vehicle control - 5% dimethyl sulfoxide, 25% polyethylene glycol 300 and 70% distilled water; 2) methyl 3,5- diphenylpyridazine-4-carboxylate at 0.1 mg/kg; 3) methyl 3,5-diphenylpyridazine-4- carboxylate at 0.3 mg/kg; 4) methyl 3,5-diphenylpyridazine-4-carboxylate at 1 mg/kg); and 5) methyl 3,5-diphenylpyridazine-4-carboxylate at 3 mg/kg; through intraperitoneal (i.p.) injection. Injections were given every other day with intervening washout days.
Each AI rat was treated with all five conditions within the testing period. To counterbalance any potential bias, drug effect was assessed using ascending-descending dose series, i.e., the dose series was given first in an ascending order and then repeated in a descending order. Therefore, each dose had two determinations.
[0275] Parametric statistics (paired t-tests) was used to compare the retention test performance of the AI rats in the two-hour delay version of the RAM task in the context of different doses of methyl 3,5-diphenylpyridazine-4-carboxylate and vehicle control (see Figure 1). The average numbers of errors that occurred in the trials were significantly fewer with methyl 3,5-diphenylpyridazine-4-carboxylate treatment of 3 mg/kg (average no. of errors ± standard error of the mean (SEM) = 1.31 ± 0.40) than using vehicle control (average no. of errors ± SEM = 3.13 ± 0.62). Relative to vehicle control treatment, methyl 3,5-diphenylpyridazine-4-carboxylate significantly improved memory performance at 3 mg/kg (t(7) = 4.233, p = 0.004).
[0276] The therapeutic dose of 3 mg/kg became ineffective when the AI rats were concurrently treated with 0.3 mg/kg of TB21007, a a5-containing GABAA R inverse agonist. The average numbers of errors made by rats with the combined TB21007/ methyl 3,5-diphenylpyridazine-4-carboxylate treatment (0.3 mg/kg TB21007 with 3 mg/kg methyl 3,5-diphenylpyridazine-4-carboxylate) was 2.88 ± 1.32, and was no different from rats treated with vehicle control (3.13 ± 1.17 average errors). Thus, the effect of methyl 3,5-diphenylpyridazine-4-carboxylate on spatial memory is a GABAA a5 receptor- dependent effect (see Figure 1).
(B) Effect of Methyl 3,5-diphenylpyridazine-4-carboxylate on a5-containing GABAA Receptor Occupancy
Animals
[0277] Adult male Long Evans rats (265-295 g, Charles River, Portage, MI, n=4/group) were used for GABAAOC5 receptor occupancy studies. Rats were individually housed in ventilated stainless-steel racks on a 12: 12 light/dark cycle. Food and water were available ad libitum. In additional studies to evaluate compound exposures at behaviorally active doses, young or aged Long Evan rats (n= 2-4/group) were used for these studies.
Compounds
[0278] Ro 15-4513 was used as a receptor occupancy (RO) tracer for GABAAOC5 receptor sites in the hippocampus and cerebellum. Ro 15-4513 was chosen as the tracer based on its selectivity for GABAAOC5 receptors relative to other alpha subunit containing GABAA receptors and because it has been successfully used for GABAAOC5 RO studies in animals and humans (see, e.g., Lingford-Hughes et ah, J. Cereb. Blood Flow Metab. 22:878-89 (2002); Pym et al, Br. J. Pharmacol. 146: 817-825 (2005); and Maeda et ah, Synapse 47: 200-208 (2003)). Ro 15-4513 (1 pg/kg), was dissolved in 25% hydroxyl- propyl beta-cyclodextrin and administered i.v. 20’ prior to the RO evaluations. Methyl 3,5-diphenylpyridazine-4-carboxylate (0.1 - 10 mg/kg) was synthesized by Nox Pharmaceuticals (India) and was dissolved in 25% hydroxyl-propyl beta-cyclodextrin and administered i.v. 15’ prior to tracer injection. Compounds were administered in a volume of 0.5 ml/kg except for the highest dose of methyl 3,5-diphenylpyridazine-4-carboxylate (10 mg/kg) which was administered in a volume of 1 ml/kg due to solubility limitations.
Tissue preparation and analysis
[0279] The rats were sacrificed by cervical dislocation 20’ post tracer injection. The whole brain was rapidly removed, and lightly rinsed with sterile water. Trunk blood was collected in EDTA coated eppendorf tubes and stored on wet ice until study completion. Hippocampus and cerebellum were dissected and stored in 1.5 ml eppendorf tubes, and placed on wet ice until tissue extraction. In a drug naive rat, six cortical brain tissues samples were collected for use in generating blank and standard curve samples.
[0280] Acetonitrile containing 0.1% formic acid was added to each sample at a volume of four times the weight of the tissue sample. For the standard curve (0.1-30 ng/g) samples, a calculated volume of standard reduced the volume of acetonitrile. The sample was homogenized (FastPrep-24, Lysing Matrix D; 5.5 m/s, for 60 seconds or 7-8 watts power using sonic probe dismembrator; Fisher Scientific) and centrifuged for 16-minutes at 14,000 rpm. The (100 pi) supernatant solution was diluted by 300 mΐ of sterile water (pH 6.5). This solution was then mixed thoroughly and analyzed via LC/MS/MS for Ro 15- 4513 (tracer) and methyl 3,5-diphenylpyridazine-4-carboxylate.
[0281] For plasma exposures, blood samples were centrifuged at 14000 rpm for 16 minutes. After centrifuging, 50ul of supernatant (plasma) from each sample was added to 200 mΐ of acetonitrile plus 0.1% formic acid. For standard curve (1-1000 ng/ml) samples, a calculated volume of standard reduced the volume of acetonitrile. Samples were sonicated for 5 minutes in an ultrasonic water bath, followed by centrifugation for 30 minutes, at 16000 RPM. lOOul of supernatant was removed from each sample vial and placed in a new glass auto sample vial, followed by the addition of 300 mΐ of sterile water (pH 6.5). This solution was then mixed thoroughly and analyzed via LC/MS/MS for methyl 3,5- diphenylpyridazine-4-carboxylate.
[0282] Receptor occupancy was determined by the ratio method which compared occupancy in the hippocampus (a region of high GABAAOC5 receptor density) with occupancy in the cerebellum (a region with low GAB AAOC5 receptor density) and additionally by a high dose of the GABAAOC5 negative allosteric modulator L-655,708 (10 mg/kg, i.v.) to define full occupancy.
[0283] Vehicle administration followed by tracer administration of 1 pg/kg, i.v., of Ro 15-4513 resulted in > 5-fold higher levels of Ro 15-4513 in hippocampus (1.93 ± 0.05 ng/g) compared with cerebellum (0.36 ± 0.02 ng/g). Methyl 3,5-diphenylpyridazine-4- carboxylate (0.01 - 10 mg/kg, i.v.) dose-dependently reduced Ro 15-4513 binding in hippocampus, without affecting cerebellum levels of Ro 15-4513 (Figure 2) with a dose of 10 mg/kg, i.v., demonstrating >90% occupancy (Figure 3). Both methods of calculating RO yielding very similar results with ED50 values for methyl 3,5- diphenylpyridazine-4-carboxylate as 1.8 mg/kg or 1.1 mg/kg based on the ratio method or using L-755,608 to define occupancy.
[0284] Methyl 3,5-diphenylpyridazine-4-carboxylate exposure was below the quantification limits (BQL) at 0.01 mg/kg, i.v., in both plasma and hippocampus and but was detectable at low levels in hippocampus at 0.1 mg/kg, i.v. (see Table 3).
Hippocampal exposure was linear as a 10-fold increase in dose from 0.1 to 1 mg/kg, i.v., resulted in a 12-fold increase in exposure. Increasing the dose from 1 to 10 mg/kg, i.v., only increased the exposure by ~5-fold. Plasma exposure increased 12-fold as the dose increased from 1 to 10 mg/kg, i.v. Table 3: % GABAA a5 Receptor Occupancy by methyl 3,5-diphenylpyridazine-4- carboxylate (0.01-10 mg/kg, i.v.). Hippocampus and Plasma Exposure of methyl 3,5- diphenylpyridazine-4-carboxylate by Treatment Group in young Long Evans rats.
[0285] Additional studies were conducted in aged Long-Evans rats in order to determine the exposures at the behavi orally relevant doses in the cognition studies. Exposure in young Long-Evans rats was also determined to bridge with the receptor occupancy studies that were conducted in young Long-Evans rats. Exposures in young and aged Long-Evans rats were relatively similar (Table 4, Figure 4). Increasing the dose 3-fold from 1 to 3 mg/kg, ip resulted in a greater than dose-proportional increase in exposure in young and aged rats in both hippocampus and plasma with increases ranging from 4.5 to 6.6-fold.
Table 4: Hippocampus and Plasma Exposure of methyl 3,5-diphenylpyridazine-4- carboxylate in Young Long Evans Rats by Treatment Group
[0286] In the RO studies, an exposure of 180 ng/g in hippocampus (1 mg/kg, i.v.) represented 32-39% receptor occupancy depending on method used to determine RO.
This exposure is comparable to that observed in aged rats at 3 mg/kg, i.p., suggesting that 30-40% RO is required for cognitive efficacy in this model.
[0287] These studies demonstrated that methyl 3,5-diphenylpyridazine-4-carboxylate produced dose-dependent increase in GABAA a.5 receptor occupancy. Methyl 3,5- diphenylpyridazine-4-carboxylate also demonstrated good brain exposure with brain/plasma ratios>l . The studies further demonstrated that methyl 3,5- diphenylpyridazine-4-carboxylate was producing its cognitive enhancing effects by positive allosteric modulation at the GABAA a.5 subtype receptor.
Example 108: Effect of Ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5- a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate in Aged-Impaired (AI) Rats
[0288] Ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3- d][l,4]diazepine-10-carboxylate, corresponding to compound number 49 in Achermann et al. Bioorg. Med. Chem. Lett., 19:5746-5752 (2009), is a selective a5-containing GABAA R agonist.
[0289] The effect of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5- a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on the in vivo spatial memory retention of aged-impaired (AI) rats was assessed in a Radial Arm Maze (RAM) behavioral task that is essentially similar to the task as described in Example 107 (A), using vehicle control (25% cyclodextrin, which was tested 3 times: at the beginning, middle and end of ascending/descending series) and six different doses levels (0.1 mg/kg, 0.3 mg/kg, 1 mg/kg, 3 mg/kg, 10 mg/kg and 30 mg/kg, each dose was tested twice) of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine- 10-carboxylate. The same experiment was repeated using the same vehicle control and doses of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3- d][l,4]diazepine-10-carboxylate, where the vehicle control was tested 5 times, the 3 mg/kg dose of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3- d][l,4]diazepine-10-carboxylate was tested 4 times, and the other doses of ethyl 3- methoxy-7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10- carboxylate were tested twice.
[0290] Parametric statistics (paired t-tests) was used to compare the retention test performance of the AI rats in the four-hour delay version of the RAM task in the context of different doses of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5- a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate and vehicle control (see Figure 5). Relative to vehicle control treatment, ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5- a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate significantly improved memory performance at 3 mg/kg (t(7) = 4.13, p = 0.004, or t(7) = 3.08, p = 0.018) and at 10 mg/kg (t(7) = 2.82, p=0.026).
[0291] The effect of ethyl 3-methoxy-7-methyl-9H-benzo[f]imidazo[l,5- a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate on a5-containing GABAA receptor occupancy was also studied following a procedure that is essentially similar to the one as described in Example 107(B) (see above). This study demonstrated that ethyl 3-methoxy- 7-methyl-9H-benzo[f]imidazo[l,5-a][l,2,4]triazolo[4,3-d][l,4]diazepine-10-carboxylate (0.01 - 10 mg/kg, i.v.) reduced Ro 15-4513 binding in hippocampus, without affecting cerebellum levels of Ro 15-4513 (Figure 6) with a dose of 10 mg/kg, i.v., demonstrating >90% occupancy (Figure 7).
Example 109: Effect of 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7- dihydro-2-benzothiophen-4(5H)-one in Aged-Impaired Rats Using a Morris Water Maze Behavioral Task
[0292] 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2- benzothiophen-4(5H)-one, corresponding to compound 44 in Chambers et al. ./. Med. Chem. 46:2227-2240 (2003) is a selective a5-containing GABAA R agonist.
[0293] The effects of 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7- dihydro-2-benzothiophen-4(5H)-one on the in vivo spatial memory retention of aged- impaired (AI) rats were assessed in a Morris water maze behavioral task. A water maze is a pool surrounded with a novel set of patterns relative to the maze. The training protocol for the water maze may be based on a modified water maze task that has been shown to be hippocampal-dependent (de Hoz et al., Eur. ./. Neurosci., 22:745-54, 2005; Steele and Morris, Hippocampus 9: 118-36, 1999).
[0294] Cognitively impaired aged rats were implanted unilaterally with a cannula into the lateral ventricle. Stereotaxic coordinates were 1.0 mm posterior to bregma, 1.5 mm lateral to midline, and 3.5 mm ventral to the skull surface. After about a week of recovery, the rats were pre-trained in a water maze for 2 days (6 trials per day) to locate a submerged escape platform hidden underneath the surface of the pool, in which the escape platform location varied from day to day. No intracerebroventricular (ICV) infusion was given during pre-training.
[0295] After pre-training, rats received ICV infusion of either 100 pg 6,6 dimethyl-3 -(3- hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one (n = 6) in 5 mΐ DMSO or vehicle DMSO (n = 5) 40 min prior to water maze training and testing. Training consisted of 8 trials per day for 2 days where the hidden escape platform remained in the same location. Rats were given 60 seconds to locate the platform with a 60 seconds inter-trial interval. The rats were given a probe test (120 seconds) 24 hr. after the end of training where the escape platform was removed. During the training, there were 4 blocks, where each block had 4 training trials.
[0296] Rats treated with vehicle and 6,6 dimethyl-3 -(3 -hydroxypropyl)thio-l-(thiazol -2- yl)-6,7-dihydro-2-benzothiophen-4(5H)-one found the escape platform about the same time at the beginning of training (block 1). In this block of training, rats treated with vehicle and 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2- benzothiophen-4(5H)-one both spent about 24 seconds to find the escape platform. However, rats treated with 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7- dihydro-2-benzothiophen-4(5H)-one were able to find the platform more proficiently (i.e., quicker) at the end of training (block 4) than those treated with vehicle alone. In block 4, rats treated with 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2- benzothiophen-4(5H)-one spent about 9.6 seconds to find the escape platform, while rats treated with vehicle spent about 19.69 seconds. These results suggest that 6,6 dimethyl-3- (3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one improved the learning of the water maze task in rats (see Figure 8(A)).
[0297] During a test trial 24 hr. after training, the escape platform was removed. The search/swim pattern of the rats was used to measure whether the rats remember where the escape platform was located during pre-trial training in order to test for the long-term memory of the rats. In this trial, “target annulus” is a designated area 1.5 times the size of the escape platform around the area where the platform was located during pre-trial training. “Opposite annulus” is a control area of the same size as the size of the target annulus, which is located opposite to the target annulus in the pool. If the rats had good long term memory, they would tend to search in the area surrounding the location where the platform was during the pre-trial training (i.e., the “target” annulus; and not the “opposite” annulus). “Time in annulus” is the amount of time in seconds that the rat spent in the target or opposite annulus area. “Number (#) of crossings” in annulus is the number of times the rat swam across the target or opposite annulus area.
[0298] Rats received vehicle spent the same amount of time in the target annulus and opposite annulus, indicating that these rats did not seem to remember where the platform was during the pre-trial training. By contrast, rats treated with 6,6 dimethyl-3 -(3- hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen-4(5H)-one spent significantly more time in the target annulus, and crossed the “target annulus” more often, as compared to the time they spent in, or the number of times they crossed the “opposite annulus”. These results suggest that 6,6 dimethyl-3 -(3 -hydroxypropyl)thio-l-(thiazol-2- yl)-6,7-dihydro-2-benzothiophen-4(5H)-one improved the long-term memory of rats in the water maze task (see, Figures 8(B) and 8(C)).
[0299] Compounds of the present invention demonstrated positive allosteric modulatory effect on the GABAA a5 receptor (See, e.g., Example 106). These compounds will enhance the effects of GABA at the GABAA a5 receptor. Therefore, compounds of the present invention should produce cognitive enhancing effects in aged-impaired animals (such as rats), similar to the effects produced by other GABAA a5 receptor selective agonists, such as methyl 3,5-diphenylpyridazine-4-carboxylate, ethyl 3-methoxy-7- methyl-9H-benzo[f]imidazo[ 1 ,5-a] [ 1 ,2,4]triazolo[4,3 -d] [ 1 ,4]diazepine- 10-carboxylate, and 6,6 dimethyl-3-(3-hydroxypropyl)thio-l-(thiazol-2-yl)-6,7-dihydro-2-benzothiophen- 4(5H)-one (See, e.g., Examples 28-30).

Claims (22)

CLAIMS We claim:
1. A compound of formula A: formula A, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein:
Y and Z are each independently selected from C and N, wherein Y and Z cannot both be
N; each occurrence of the bond is either a single bond or a double bond; each R1 is independently halogen, -OH, or -0(C1-C6)alkyl; each R2 is -H, -OR8, -SR8, -(CH2)nOR8, -(CH2)nSR8; each of R9 is -H, (C6-C12)aryl, or 5- to 10-membered heteroaryl, wherein each R9 is substituted with 0-5 R11; each occurrence of R11 is independently selected from -halogen, -CF3, -OH, -OCF3,
OCHF2, -0-(Cl-C6)alkyl, -CN, -SCH3 -(C6-C10)aryl, and -(Cl-C6)alkyl; and m and n are independently integers selected from 0-4.
2. The compound according to claim 1, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, wherein: each R1 is halogen or -OMe; each R2 is -H or -CH2OMe; each , wherein each R9 is substituted with 0-5 R11; and each occurrence of R , nu is independently selected from -halogen, -CF3, -OH, -OCF3, OCHF2, or -OMe.
3. The compound according to claim 1 or 2, wherein the compound has a structure according to formula B: formula B, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
4. The compound according to claim 1 or 2, wherein the compound has a structure according to formula C: formula C, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
5. A compound selected from:
or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof.
6. A pharmaceutical composition comprising a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, hydrate, solvate, polymorph, isomer, or combination thereof, in a therapeutically effective amount; and an acceptable carrier, adjuvant or vehicle.
7. The pharmaceutical composition according to claim 6, wherein said composition further comprises a second therapeutic agent.
8 The pharmaceutical composition according to claim 7, wherein the second therapeutic agent is selected from an antipsychotic, memantine and an acetylcholine esterase inhibitor (AChE-I).
9. The pharmaceutical composition according to claim 7, wherein the second therapeutic agent is an antipsychotic selected from aripiprazole, olanzapine and ziprasidone, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
10. The pharmaceutical composition according to claim 7, wherein the second therapeutic agent is memantine, a pharmaceutically acceptable salt, hydrate, solvate, or polymorph thereof.
11. The pharmaceutical composition according to claim 8, wherein the second therapeutic agent is an AChE-I selected from Donepezil, Galantamine, and Rivastigmine, and the pharmaceutically acceptable salts, hydrates, solvates, and polymorphs thereof.
12. A method of treating cognitive impairment associated with a central nervous system (CNS) disorder in a subject in need thereof, comprising the step of administering a compound according to any one of claims 1-5 or a pharmaceutical composition according to any one of claims 6-11.
13. The method according to claim 12, wherein the CNS disorder is age-related cognitive impairment.
14. The method according to claim 12, wherein the cognitive impairment is Mild Cognitive Impairment (MCI).
15. The method according to claim 14, wherein the Mild Cognitive Impairment is amnestic Mild Cognitive Impairment (AMCI).
16. The method according to claim 12, wherein the CNS disorder is dementia.
17. The method according to claim 16, wherein the dementia is Alzheimer’s disease.
18. The method according to claim 12, wherein the CNS disorder is schizophrenia, amyotrophic lateral sclerosis (ALS), post-traumatic stress disorder (PTSD), mental retardation, Parkinson’s disease (PD), autism, compulsive behavior, substance addiction, bipolar disorder, or a disorder associated with cancer therapy.
19. A method of treating a brain cancer in a subject in need thereof, comprising the step of administering a compound according to any one of claims 1-5, or a pharmaceutical composition according to any one of claims 6-11.
20. A method of treating cognitive impairment associated with a brain cancer in a subject in need thereof, comprising the step of administering a compound according to any one of claims 1-5, or a pharmaceutical composition according to any one of claims 6-11.
21. The method according to claim 19 or 20, wherein said brain cancer is medulloblastoma.
22. A method of treating Parkinson’s disease psychosis in a patient in need thereof, comprising the step of administering a compound according to any one of claims 1-5, or a pharmaceutical composition according to any one of claims 6-11.
AU2020405233A 2019-12-19 2020-12-18 Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment Pending AU2020405233A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201962950886P 2019-12-19 2019-12-19
US62/950,886 2019-12-19
PCT/US2020/066182 WO2021127543A1 (en) 2019-12-19 2020-12-18 Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment

Publications (1)

Publication Number Publication Date
AU2020405233A1 true AU2020405233A1 (en) 2022-07-14

Family

ID=76478828

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2020405233A Pending AU2020405233A1 (en) 2019-12-19 2020-12-18 Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment

Country Status (8)

Country Link
US (1) US20230098667A1 (en)
EP (1) EP4077333A4 (en)
JP (1) JP2023507569A (en)
CN (1) CN115175911A (en)
AU (1) AU2020405233A1 (en)
IL (1) IL294031A (en)
MX (1) MX2022007725A (en)
WO (1) WO2021127543A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220246017A1 (en) * 2019-05-23 2022-08-04 Life Guard Electrooptic Ltd. System and a method for surveilling swimmers
WO2024023268A1 (en) 2022-07-29 2024-02-01 F. Hoffmann-La Roche Ag Characterisation of neurological dysfunction
WO2024039886A1 (en) * 2022-08-19 2024-02-22 Agenebio, Inc. Benzazepine derivatives, compositions, and methods for treating cognitive impairment

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106068256B (en) * 2013-12-20 2020-10-23 艾吉因生物股份有限公司 Benzodiazepine derivatives, compositions and methods for treating cognitive impairment
EP3310785A4 (en) * 2015-06-19 2018-12-26 Agenebio, Inc. Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US20180170941A1 (en) * 2016-12-19 2018-06-21 Agenebio, Inc. Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
CA3104478A1 (en) * 2018-06-19 2019-12-26 Agenebio, Inc. Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
CA3185573A1 (en) * 2020-07-10 2022-01-13 Deepa GANDLA Polymorphs of a gabaa .alpha.5 agonist and methods of using in the treatment of cognitive impairment
US20230270753A1 (en) * 2020-07-10 2023-08-31 Agenebio, Inc. Combinations of gabaa alpha 5 agonists and sv2a inhibitors and methods of using in the treatment of cognitive impairment

Also Published As

Publication number Publication date
CN115175911A (en) 2022-10-11
JP2023507569A (en) 2023-02-24
MX2022007725A (en) 2022-07-19
US20230098667A1 (en) 2023-03-30
EP4077333A4 (en) 2024-01-10
EP4077333A1 (en) 2022-10-26
WO2021127543A1 (en) 2021-06-24
IL294031A (en) 2022-08-01

Similar Documents

Publication Publication Date Title
US11142529B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US20230134844A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US11414425B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US20210009602A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US20230098667A1 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US11505555B2 (en) Benzodiazepine derivatives, compositions, and methods for treating cognitive impairment
US20240132513A1 (en) Benzazepine derivatives, compositions, and methods for treating cognitive impairment
WO2024039886A1 (en) Benzazepine derivatives, compositions, and methods for treating cognitive impairment